1
|
Demler C, Lawlor JC, Yelin R, Llivichuzcha-Loja D, Shaulov L, Kim D, Stewart M, Lee FK, Shylo N, Trainor PA, Schultheiss TM, Kurpios NA. An atypical basement membrane forms a midline barrier during left-right asymmetric gut development in the chicken embryo. eLife 2025; 12:RP89494. [PMID: 40298919 PMCID: PMC12040318 DOI: 10.7554/elife.89494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Correct intestinal morphogenesis depends on the early embryonic process of gut rotation, an evolutionarily conserved program in which a straight gut tube elongates and forms into its first loops. However, the gut tube requires guidance to loop in a reproducible manner. The dorsal mesentery (DM) connects the gut tube to the body and directs the lengthening gut into stereotypical loops via left-right (LR) asymmetric cellular and extracellular behavior. The LR asymmetry of the DM also governs blood and lymphatic vessel formation for the digestive tract, which is essential for prenatal organ development and postnatal vital functions including nutrient absorption. Although the genetic LR asymmetry of the DM has been extensively studied, a divider between the left and right DM has yet to be identified. Setting up LR asymmetry for the entire body requires a Lefty1+ midline barrier to separate the two sides of the embryo, without it, embryos have lethal or congenital LR patterning defects. Individual organs including the brain, heart, and gut also have LR asymmetry, and while the consequences of left and right signals mixing are severe or even lethal, organ-specific mechanisms for separating these signals remain poorly understood. Here, we uncover a midline structure composed of a transient double basement membrane, which separates the left and right halves of the embryonic chick DM during the establishment of intestinal and vascular asymmetries. Unlike other basement membranes of the DM, the midline is resistant to disruption by intercalation of Netrin4 (Ntn4). We propose that this atypical midline forms the boundary between left and right sides and functions as a barrier necessary to establish and protect organ asymmetry.
Collapse
Affiliation(s)
- Cora Demler
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - John C Lawlor
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
| | - Dhana Llivichuzcha-Loja
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Lihi Shaulov
- Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
| | - David Kim
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Megan Stewart
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Frank K Lee
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Natalia Shylo
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Paul A Trainor
- Stowers Institute for Medical ResearchKansas CityUnited States
- Department of Anatomy and Cell Biology, University of Kansas Medical CenterKansas CityUnited States
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| |
Collapse
|
2
|
Zhou N, Zhang S, Wang C, Zheng B, Zhang A, Zhou W. Generation of human induced pluripotent stem cell lines derived from a patient carrying an intragenic deletion in the NFIA gene. Hum Cell 2025; 38:95. [PMID: 40266456 DOI: 10.1007/s13577-025-01222-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025]
Abstract
Brain malformations with or without urinary tract defects (BRMUTD) are caused by heterozygous variants in the NFIA gene. BRMUTD is a neurodevelopmental disorder characterized by hypoplasia or absence of the corpus callosum, hydrocephalus or ventriculomegaly, and developmental delay, which may or may not be accompanied by urinary tract defects. Here, we report the successful generation of induced pluripotent stem cells (hiPSCs) from a 3-year-old male BRMUTD patient using Sendai virus-based non-integrating reprogramming technology. This patient-derived cell line harbors an intragenic deletion within the NFIA gene (NC_000001.10: g.61650967_61842967del [GRCh37]), which is associated with a significant reduction in NFIA expression. This cell line maintains a normal karyotype, expresses pluripotency markers, and can differentiate into three germ layers. The established hiPSCs line will provide an in vitro model for studying pathological mechanisms and potential therapies of NFIA-related neurodevelopmental disorder.
Collapse
Affiliation(s)
- Ning Zhou
- School of Medicine, Southeast University, Nanjing, 210009, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Shengnan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Chunli Wang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Aihua Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| | - Wei Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| |
Collapse
|
3
|
Esteve NA, Rogers DJ, Stagray JA, Mayeux H, Nora G, Huval L, Smith KM. Tanycyte radial morphology and proliferation are influenced by fibroblast growth factor receptor 1 and high-fat diet. Eur J Neurosci 2024; 60:5000-5018. [PMID: 39087621 DOI: 10.1111/ejn.16473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 04/19/2024] [Accepted: 07/07/2024] [Indexed: 08/02/2024]
Abstract
Fibroblast growth factor receptor 1 (FGFR1) is a widely expressed, membrane-bound receptor that transduces extracellular signals from FGF ligands and cadherins, resulting in intracellular signals influencing cellular growth, proliferation, calcium, and transcription. FGF21 and FGF2 stimulate the proliferation of tanycytes, specialized radial astrocytes along the ventricle of the hypothalamus, and influence metabolism. Tanycytes are in a privileged position between the cerebrospinal fluid, the blood supply in the median eminence, and neurons within nuclei in the hypothalamus. The effect of FGFR1 signaling upon tanycyte morphology and metabolism was examined in adult mice with conditional deletion of the Fgfr1 gene using the Fgfr1flox/flox; Nestin-Cre+ line. Loss of Fgfr1 resulted in shorter β tanycytes along the medial eminence. Control Fgfr1flox/flox littermates and Fgfr1flox/flox, Nestin-Cre+ (Fgfr1 cKO) knockout mice were placed on a 1-month long high-fat diet (HFD) or a normal-fat diet (NFD), to investigate differences in body homeostasis and tanycyte morphology under an obesity inducing diet. We found that FGFR1 is a vital contributor to tanycyte morphology and quantity and that it promotes stem cell maintenance in the hypothalamus and hippocampal dentate gyrus. The Fgfr1 cKO mice developed impaired tolerance to a glucose challenge test on a HFD without gaining more weight than control mice. The combination of HFD and loss of Fgfr1 gene resulted in altered β and α tanycyte morphology, and reduced stem cell numbers along the third ventricle of the hypothalamus and hippocampus.
Collapse
Affiliation(s)
- N Alex Esteve
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisianafs, USA
| | - Deborah J Rogers
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisianafs, USA
| | - Jacob A Stagray
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisianafs, USA
| | - Holly Mayeux
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisianafs, USA
| | - Glenae Nora
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisianafs, USA
| | - Luke Huval
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisianafs, USA
| | - Karen Müller Smith
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisianafs, USA
| |
Collapse
|
4
|
Demler C, Lawlor JC, Yelin R, Llivichuzcha-Loja D, Shaulov L, Kim D, Stewart M, Lee F, Shylo NA, Trainor PA, Schultheiss T, Kurpios NA. An atypical basement membrane forms a midline barrier during left-right asymmetric gut development in the chicken embryo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.15.553395. [PMID: 37645918 PMCID: PMC10461973 DOI: 10.1101/2023.08.15.553395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Correct intestinal morphogenesis depends on the early embryonic process of gut rotation, an evolutionarily conserved program in which a straight gut tube elongates and forms into its first loops. However, the gut tube requires guidance to loop in a reproducible manner. The dorsal mesentery (DM) connects the gut tube to the body and directs the lengthening gut into stereotypical loops via left-right (LR) asymmetric cellular and extracellular behavior. The LR asymmetry of the DM also governs blood and lymphatic vessel formation for the digestive tract, which is essential for prenatal organ development and postnatal vital functions including nutrient absorption. Although the genetic LR asymmetry of the DM has been extensively studied, a divider between the left and right DM has yet to be identified. Setting up LR asymmetry for the entire body requires a Lefty1+ midline barrier to separate the two sides of the embryo, without it, embryos have lethal or congenital LR patterning defects. Individual organs including the brain, heart, and gut also have LR asymmetry, and while the consequences of left and right signals mixing are severe or even lethal, organ-specific mechanisms for separating these signals are poorly understood. Here, we uncover a midline structure composed of a transient double basement membrane, which separates the left and right halves of the embryonic chick DM during the establishment of intestinal and vascular asymmetries. Unlike other basement membranes of the DM, the midline is resistant to disruption by intercalation of Netrin4 (Ntn4). We propose that this atypical midline forms the boundary between left and right sides and functions as a barrier necessary to establish and protect organ asymmetry.
Collapse
Affiliation(s)
- Cora Demler
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - John Coates Lawlor
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Dhana Llivichuzcha-Loja
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Lihi Shaulov
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - David Kim
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Megan Stewart
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Missouri, USA
| | - Thomas Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Natasza A. Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
5
|
Ye Y, Jin B, Zhang HW, Sheng N. Strategies for Dissecting the Genetic Driving of Conserved Noncoding-Elements for Evolutionary Development of the Corpus Callosum. Neurosci Bull 2024; 40:1025-1027. [PMID: 38568390 PMCID: PMC11251079 DOI: 10.1007/s12264-024-01198-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/13/2024] [Indexed: 07/16/2024] Open
Affiliation(s)
- Yaxin Ye
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650201, China
| | - Boxing Jin
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650201, China
| | - Hao W Zhang
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650201, China
| | - Nengyin Sheng
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650201, China.
| |
Collapse
|
6
|
Martins-Costa C, Wiegers A, Pham VA, Sidhaye J, Doleschall B, Novatchkova M, Lendl T, Piber M, Peer A, Möseneder P, Stuempflen M, Chow SYA, Seidl R, Prayer D, Höftberger R, Kasprian G, Ikeuchi Y, Corsini NS, Knoblich JA. ARID1B controls transcriptional programs of axon projection in an organoid model of the human corpus callosum. Cell Stem Cell 2024; 31:866-885.e14. [PMID: 38718796 DOI: 10.1016/j.stem.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 02/13/2024] [Accepted: 04/17/2024] [Indexed: 06/09/2024]
Abstract
Mutations in ARID1B, a member of the mSWI/SNF complex, cause severe neurodevelopmental phenotypes with elusive mechanisms in humans. The most common structural abnormality in the brain of ARID1B patients is agenesis of the corpus callosum (ACC), characterized by the absence of an interhemispheric white matter tract that connects distant cortical regions. Here, we find that neurons expressing SATB2, a determinant of callosal projection neuron (CPN) identity, show impaired maturation in ARID1B+/- neural organoids. Molecularly, a reduction in chromatin accessibility of genomic regions targeted by TCF-like, NFI-like, and ARID-like transcription factors drives the differential expression of genes required for corpus callosum (CC) development. Through an in vitro model of the CC tract, we demonstrate that this transcriptional dysregulation impairs the formation of long-range axonal projections, causing structural underconnectivity. Our study uncovers new functions of the mSWI/SNF during human corticogenesis, identifying cell-autonomous axonogenesis defects in SATB2+ neurons as a cause of ACC in ARID1B patients.
Collapse
Affiliation(s)
- Catarina Martins-Costa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Andrea Wiegers
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Vincent A Pham
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Jaydeep Sidhaye
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Balint Doleschall
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Maria Novatchkova
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Thomas Lendl
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Marielle Piber
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Angela Peer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Paul Möseneder
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Marlene Stuempflen
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Siu Yu A Chow
- Institute of Industrial Science, The University of Tokyo, 153-8505 Tokyo, Japan; Institute for AI and Beyond, The University of Tokyo, 113-0032 Tokyo, Japan
| | - Rainer Seidl
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniela Prayer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gregor Kasprian
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Yoshiho Ikeuchi
- Institute of Industrial Science, The University of Tokyo, 153-8505 Tokyo, Japan; Institute for AI and Beyond, The University of Tokyo, 113-0032 Tokyo, Japan
| | - Nina S Corsini
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria.
| | - Jürgen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria; Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
7
|
Mitchell KJ. Variability in Neural Circuit Formation. Cold Spring Harb Perspect Biol 2024; 16:a041504. [PMID: 38253418 PMCID: PMC10910361 DOI: 10.1101/cshperspect.a041504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The study of neural development is usually concerned with the question of how nervous systems get put together. Variation in these processes is usually of interest as a means of revealing these normative mechanisms. However, variation itself can be an object of study and is of interest from multiple angles. First, the nature of variation in both the processes and the outcomes of neural development is relevant to our understanding of how these processes and outcomes are encoded in the genome. Second, variation in the wiring of the brain in humans may underlie variation in all kinds of psychological and behavioral traits, as well as neurodevelopmental disorders. And third, genetic variation that affects circuit development provides the raw material for evolutionary change. Here, I examine these different aspects of variation in circuit development and consider what they may tell us about these larger questions.
Collapse
Affiliation(s)
- Kevin J Mitchell
- Smurfit Institute of Genetics and Institute of Neuroscience, Trinity College Dublin, Dublin D02 PN40, Ireland
| |
Collapse
|
8
|
Rodríguez-Pérez LM, López-de-San-Sebastián J, de Diego I, Smith A, Roales-Buján R, Jiménez AJ, Paez-Gonzalez P. A selective defect in the glial wedge as part of the neuroepithelium disruption in hydrocephalus development in the mouse hyh model is associated with complete corpus callosum dysgenesis. Front Cell Neurosci 2024; 18:1330412. [PMID: 38450283 PMCID: PMC10915275 DOI: 10.3389/fncel.2024.1330412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Dysgenesis of the corpus callosum is present in neurodevelopmental disorders and coexists with hydrocephalus in several human congenital syndromes. The mechanisms that underlie the etiology of congenital hydrocephalus and agenesis of the corpus callosum when they coappear during neurodevelopment persist unclear. In this work, the mechanistic relationship between both disorders is investigated in the hyh mouse model for congenital hydrocephalus, which also develops agenesis of the corpus callosum. In this model, hydrocephalus is generated by a defective program in the development of neuroepithelium during its differentiation into radial glial cells. Methods In this work, the populations implicated in the development of the corpus callosum (callosal neurons, pioneering axons, glial wedge cells, subcallosal sling and indusium griseum glial cells) were studied in wild-type and hyh mutant mice. Immunohistochemistry, mRNA in situ hybridization, axonal tracing experiments, and organotypic cultures from normal and hyh mouse embryos were used. Results Our results show that the defective program in the neuroepithelium/radial glial cell development in the hyh mutant mouse selectively affects the glial wedge cells. The glial wedge cells are necessary to guide the pioneering axons as they approach the corticoseptal boundary. Our results show that the pioneering callosal axons arising from neurons in the cingulate cortex can extend projections to the interhemispheric midline in normal and hyh mice. However, pioneering axons in the hyh mutant mouse, when approaching the area corresponding to the damaged glial wedge cell population, turned toward the ipsilateral lateral ventricle. This defect occurred before the appearance of ventriculomegaly. Discussion In conclusion, the abnormal development of the ventricular zone, which appears to be inherent to the etiology of several forms of congenital hydrocephalus, can explain, in some cases, the common association between hydrocephalus and corpus callosum dysgenesis. These results imply that further studies may be needed to understand the corpus callosum dysgenesis etiology when it concurs with hydrocephalus.
Collapse
Affiliation(s)
- Luis-Manuel Rodríguez-Pérez
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Universidad de Málaga, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
| | | | - Isabel de Diego
- Departamento de Anatomía y Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Malaga, Spain
| | - Aníbal Smith
- Departamento de Anatomía y Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Malaga, Spain
| | - Ruth Roales-Buján
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| | - Antonio J. Jiménez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| | - Patricia Paez-Gonzalez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| |
Collapse
|
9
|
Lynton Z, Suárez R, Fenlon LR. Brain plasticity following corpus callosum agenesis or loss: a review of the Probst bundles. Front Neuroanat 2023; 17:1296779. [PMID: 38020213 PMCID: PMC10657877 DOI: 10.3389/fnana.2023.1296779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
The corpus callosum is the largest axonal tract in the human brain, connecting the left and right cortical hemipheres. This structure is affected in myriad human neurodevelopmental disorders, and can be entirely absent as a result of congenital or surgical causes. The age when callosal loss occurs, for example via surgical section in cases of refractory epilepsy, correlates with resulting brain morphology and neuropsychological outcomes, whereby an earlier loss generally produces relatively improved interhemispheric connectivity compared to a loss in adulthood (known as the "Sperry's paradox"). However, the mechanisms behind these age-dependent differences remain unclear. Perhaps the best documented and most striking of the plastic changes that occur due to developmental, but not adult, callosal loss is the formation of large, bilateral, longitudinal ectopic tracts termed Probst bundles. Despite over 100 years of research into these ectopic tracts, which are the largest and best described stereotypical ectopic brain tracts in humans, much remains unclear about them. Here, we review the anatomy of the Probst bundles, along with evidence for their faciliatory or detrimental function, the required conditions for their formation, patterns of etiology, and mechanisms of development. We provide hypotheses for many of the remaining mysteries of the Probst bundles, including their possible relationship to preserved interhemispheric communication following corpus callosum absence. Future research into naturally occurring plastic tracts such as Probst bundles will help to inform the general rules governing axon plasticity and disorders of brain miswiring.
Collapse
Affiliation(s)
- Zorana Lynton
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Rodrigo Suárez
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Laura R. Fenlon
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
10
|
Yan K, Bormuth I, Bormuth O, Tutukova S, Renner A, Bessa P, Schaub T, Rosário M, Tarabykin V. TrkB-dependent EphrinA reverse signaling regulates callosal axon fasciculate growth downstream of Neurod2/6. Cereb Cortex 2023; 33:1752-1767. [PMID: 35462405 DOI: 10.1093/cercor/bhac170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/14/2022] Open
Abstract
Abnormal development of corpus callosum is relatively common and causes a broad spectrum of cognitive impairments in humans. We use acallosal Neurod2/6-deficient mice to study callosal axon guidance within the ipsilateral cerebral cortex. Initial callosal tracts form but fail to traverse the ipsilateral cingulum and are not attracted towards the midline in the absence of Neurod2/6. We show that the restoration of Ephrin-A4 (EfnA4) expression in the embryonic neocortex of Neurod2/6-deficient embryos is sufficient to partially rescue targeted callosal axon growth towards the midline. EfnA4 cannot directly mediate reverse signaling within outgrowing axons, but it forms co-receptor complexes with TrkB (Ntrk2). The ability of EfnA4 to rescue the guided growth of a subset of callosal axons in Neurod2/6-deficient mice is abolished by the co-expression of dominant negative TrkBK571N (kinase-dead) or TrkBY515F (SHC-binding deficient) variants, but not by TrkBY816F (PLCγ1-binding deficient). Additionally, EphA4 is repulsive to EfnA4-positive medially projecting axons in organotypic brain slice culture. Collectively, we suggest that EfnA4-mediated reverse signaling acts via TrkB-SHC and is required for ipsilateral callosal axon growth accuracy towards the midline downstream of Neurod family factors.
Collapse
Affiliation(s)
- Kuo Yan
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Ingo Bormuth
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Olga Bormuth
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany.,Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia
| | - Svetlana Tutukova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia.,Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634009, Tomsk, Russia
| | - Ana Renner
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Paraskevi Bessa
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Theres Schaub
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Marta Rosário
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany.,Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia.,Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634009, Tomsk, Russia
| |
Collapse
|
11
|
Gao G, Hausmann S, Flores NM, Benitez AM, Shen J, Yang X, Person MD, Gayatri S, Cheng D, Lu Y, Liu B, Mazur PK, Bedford MT. The NFIB/CARM1 partnership is a driver in preclinical models of small cell lung cancer. Nat Commun 2023; 14:363. [PMID: 36690626 PMCID: PMC9870865 DOI: 10.1038/s41467-023-35864-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
The coactivator associated arginine methyltransferase (CARM1) promotes transcription, as its name implies. It does so by modifying histones and chromatin bound proteins. We identified nuclear factor I B (NFIB) as a CARM1 substrate and show that this transcription factor utilizes CARM1 as a coactivator. Biochemical studies reveal that tripartite motif 29 (TRIM29) is an effector molecule for methylated NFIB. Importantly, NFIB harbors both oncogenic and metastatic activities, and is often overexpressed in small cell lung cancer (SCLC). Here, we explore the possibility that CARM1 methylation of NFIB is important for its transforming activity. Using a SCLC mouse model, we show that both CARM1 and the CARM1 methylation site on NFIB are critical for the rapid onset of SCLC. Furthermore, CARM1 and methylated NFIB are responsible for maintaining similar open chromatin states in tumors. Together, these findings suggest that CARM1 might be a therapeutic target for SCLC.
Collapse
Affiliation(s)
- Guozhen Gao
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Simone Hausmann
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Natasha M Flores
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ana Morales Benitez
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaojie Yang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maria D Person
- Center for Biomedical Research Support, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sitaram Gayatri
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Evozyne Inc., Chicago, IL, 60614, USA
| | - Donghang Cheng
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bin Liu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Pawel K Mazur
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Mark T Bedford
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Moradi B, Taherian R, Tahmasebpour AR, Sanei Taheri M, Kazemi MA, Pak N, Shirazi M, Radmanesh A, Oztekin O, Arab-Ahmadi M. Fetal corpus callosum abnormalities: Ultrasound and magnetic resonance imaging role. JOURNAL OF CLINICAL ULTRASOUND : JCU 2022; 50:989-1003. [PMID: 35488776 DOI: 10.1002/jcu.23212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 06/14/2023]
Abstract
The corpus callosum (CC) is the major interhemispheric commissure and its abnormalities include agenesis, hypoplasia, and hyperplasia. The CC anomalies are typically related to other central nervous system (CNS) or extra-CNS malformations. The antenatal diagnosis of complete CC agenesis is easy after mid-trimester by ultrasound (US) even in the axial plane. The non-visualization of cavum septum pellucidum and colpocephaly are critical signs in the axial view. More subtle findings (i.e., hypoplasia and partial agenesis) might also be recognized antenatally. In this review, the focus was given on the prenatal diagnosis of CC abnormalities in US and magnetic resonance imaging.
Collapse
Affiliation(s)
- Behnaz Moradi
- Department of Radiology, Yas Complex Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Taherian
- Department of Radiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Morteza Sanei Taheri
- Department of Radiology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Kazemi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiology, Amir Alam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Pak
- Department of Radiology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboobeh Shirazi
- Maternal Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Radmanesh
- Department of Radiology, School of Medicine, New York University, New York, New York, USA
| | - Ozgur Oztekin
- Radiology Department, Izmir Education and Research Hospital, Izmir, Turkey
| | - Mehran Arab-Ahmadi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Ezan J, Moreau MM, Mamo TM, Shimbo M, Decroo M, Sans N, Montcouquiol M. Neuron-Specific Deletion of Scrib in Mice Leads to Neuroanatomical and Locomotor Deficits. Front Genet 2022; 13:872700. [PMID: 35692812 PMCID: PMC9174639 DOI: 10.3389/fgene.2022.872700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Scribble (Scrib) is a conserved polarity protein acting as a scaffold involved in multiple cellular and developmental processes. Recent evidence from our group indicates that Scrib is also essential for brain development as early global deletion of Scrib in the dorsal telencephalon induced cortical thickness reduction and alteration of interhemispheric connectivity. In addition, Scrib conditional knockout (cKO) mice have behavioral deficits such as locomotor activity impairment and memory alterations. Given Scrib broad expression in multiple cell types in the brain, we decided to determine the neuronal contribution of Scrib for these phenotypes. In the present study, we further investigate the function of Scrib specifically in excitatory neurons on the forebrain formation and the control of locomotor behavior. To do so, we generated a novel neuronal glutamatergic specific Scrib cKO mouse line called Nex-Scrib−/− cKO. Remarkably, cortical layering and commissures were impaired in these mice and reproduced to some extent the previously described phenotype in global Scrib cKO. In addition and in contrast to our previous results using Emx1-Scrib−/− cKO, the Nex-Scrib−/− cKO mutant mice exhibited significantly reduced locomotion. Altogether, the novel cKO model described in this study further highlights an essential role for Scrib in forebrain development and locomotor behavior.
Collapse
Affiliation(s)
- Jerome Ezan
- INSERM U1215, Neurocentre Magendie, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, INSERM U1215, F-33000, Bordeaux, France
- *Correspondence: Jerome Ezan,
| | - Maité M. Moreau
- INSERM U1215, Neurocentre Magendie, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, INSERM U1215, F-33000, Bordeaux, France
| | - Tamrat M. Mamo
- INSERM U1215, Neurocentre Magendie, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, INSERM U1215, F-33000, Bordeaux, France
| | - Miki Shimbo
- INSERM U1215, Neurocentre Magendie, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, INSERM U1215, F-33000, Bordeaux, France
| | - Maureen Decroo
- INSERM U1215, Neurocentre Magendie, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, INSERM U1215, F-33000, Bordeaux, France
| | - Nathalie Sans
- INSERM U1215, Neurocentre Magendie, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, INSERM U1215, F-33000, Bordeaux, France
| | - Mireille Montcouquiol
- INSERM U1215, Neurocentre Magendie, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, INSERM U1215, F-33000, Bordeaux, France
| |
Collapse
|
14
|
Pediatric Brain Maturation and Migration Disorders. Diagnostics (Basel) 2022; 12:diagnostics12051123. [PMID: 35626279 PMCID: PMC9139849 DOI: 10.3390/diagnostics12051123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Neurodevelopmental disorders, including neuronal migration disorders, are best understood in the context of altered normal development. Neurons normally migrate from their site of origin to their (usually cortical) destination using a wide range of molecular and cellular signaling as a guide. In the case of abnormal migration neurons: (1) do not migrate and remain at their site of origin; (2) incompletely migrate and remain within the white matter; (3) migrate to the cortex but fail to organize correctly; or (4) over-migrate, beyond the cortex. In this review, we discuss normal brain development, along with the malformations that result from these different migration abnormalities.
Collapse
|
15
|
Kozulin P, Suárez R, Zhao QY, Paolino A, Richards LJ, Fenlon LR. Divergent evolution of developmental timing in the neocortex revealed by marsupial and eutherian transcriptomes. Development 2022; 149:274319. [DOI: 10.1242/dev.200212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022]
Abstract
ABSTRACT
Only mammals evolved a neocortex, which integrates sensory-motor and cognitive functions. Significant diversifications in the cellular composition and connectivity of the neocortex occurred between the two main therian groups: marsupials and eutherians. However, the developmental mechanisms underlying these diversifications are largely unknown. Here, we compared the neocortical transcriptomes of Sminthopsis crassicaudata, a mouse-sized marsupial, with those of eutherian mice at two developmentally equivalent time points corresponding to deeper and upper layer neuron generation. Enrichment analyses revealed more mature gene networks in marsupials at the early stage, which reverted at the later stage, suggesting a more precocious but protracted neuronal maturation program relative to birth timing of cortical layers. We ranked genes expressed in different species and identified important differences in gene expression rankings between species. For example, genes known to be enriched in upper-layer cortical projection neuron subtypes, such as Cux1, Lhx2 and Satb2, likely relate to corpus callosum emergence in eutherians. These results show molecular heterochronies of neocortical development in Theria, and highlight changes in gene expression and cell type composition that may underlie neocortical evolution and diversification.
This article has an associated ‘The people behind the papers’ interview.
Collapse
Affiliation(s)
- Peter Kozulin
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Rodrigo Suárez
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Qiong-Yi Zhao
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Annalisa Paolino
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Linda J. Richards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Laura R. Fenlon
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
16
|
Ogura Y, Uehara T, Ujibe K, Yoshihashi H, Yamada M, Suzuki H, Takenouchi T, Kosaki K, Hirata H. The p.Thr395Met missense variant of NFIA found in a patient with intellectual disability is a defective variant. Am J Med Genet A 2022; 188:1184-1192. [PMID: 35018717 DOI: 10.1002/ajmg.a.62638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/05/2021] [Accepted: 12/11/2021] [Indexed: 12/17/2022]
Abstract
Nuclear factor one A (NFIA) is a transcription factor that regulates the development of the central nervous system. Haploinsufficiency of the NFIA gene causes NFIA-related disorder, which includes brain abnormalities and intellectual disability, with or without urinary tract defects. Intragenic deletions, nonsense variants, frameshift variants, and missense variants in one allele of the NFIA gene have been reported to cause various neurological and urogenital symptoms. Here we report a 10-year-old male patient with developmental delay, coarctation of the aorta, and distinctive facial features. Exome analysis identified a rare de novo heterozygous missense variant p.Thr395Met in NFIA. We employed zebrafish as a model organism in our NFIA analysis and found that nfia-/- zebrafish initially showed a loss of commissural axons in the brain, and eventually underwent growth retardation resulting in premature death. Impairment of the commissural neurons in nfia-/- zebrafish embryos could be restored by the expression of wild-type human NFIA protein, but not of mutant human protein harboring the p.Thr395Met substitution, indicating that this variant affects the function of NFIA protein. Taken together, we suggest that the p.Thr395Met allele in the NFIA gene is relevant to the pathogenesis of NFIA-related disorder.
Collapse
Affiliation(s)
- Yurie Ogura
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University, Sagamihara, Japan
| | - Tomoko Uehara
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan.,Department of Clinical Genetics, Aichi Developmental Disability Center Central Hospital, Aichi, Japan
| | - Kota Ujibe
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University, Sagamihara, Japan
| | - Hiroshi Yoshihashi
- Department of Clinical Genetics, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Mamiko Yamada
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Hisato Suzuki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Toshiki Takenouchi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Hiromi Hirata
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University, Sagamihara, Japan
| |
Collapse
|
17
|
Ahmed G, Shinmyo Y. Multiple Functions of Draxin/Netrin-1 Signaling in the Development of Neural Circuits in the Spinal Cord and the Brain. Front Neuroanat 2021; 15:766911. [PMID: 34899198 PMCID: PMC8655782 DOI: 10.3389/fnana.2021.766911] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/22/2021] [Indexed: 11/30/2022] Open
Abstract
Axon guidance proteins play key roles in the formation of neural circuits during development. We previously identified an axon guidance cue, named draxin, that has no homology with other axon guidance proteins. Draxin is essential for the development of various neural circuits including the spinal cord commissure, corpus callosum, and thalamocortical projections. Draxin has been shown to not only control axon guidance through netrin-1 receptors, deleted in colorectal cancer (Dcc), and neogenin (Neo1) but also modulate netrin-1-mediated axon guidance and fasciculation. In this review, we summarize the multifaceted functions of draxin and netrin-1 signaling in neural circuit formation in the central nervous system. Furthermore, because recent studies suggest that the distributions and functions of axon guidance cues are highly regulated by glycoproteins such as Dystroglycan and Heparan sulfate proteoglycans, we discuss a possible function of glycoproteins in draxin/netrin-1-mediated axon guidance.
Collapse
Affiliation(s)
- Giasuddin Ahmed
- Department of Neuroscience and Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
18
|
Rayêe D, Iack PM, Christoff RR, Lourenço MR, Bonifácio C, Boltz J, Lent R, Garcez PP. The Dynamics of Axon Bifurcation Development in the Cerebral Cortex of Typical and Acallosal Mice. Neuroscience 2021; 477:14-24. [PMID: 34601063 DOI: 10.1016/j.neuroscience.2021.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 11/30/2022]
Abstract
The corpus callosum (CC) is a major interhemispheric commissure of placental mammals. Early steps of CC formation rely on guidance strategies, such as axonal branching and collateralization. Here we analyze the time-course dynamics of axonal bifurcation during typical cortical development or in a CC dysgenesis mouse model. We use Swiss mice as a typical CC mouse model and find that axonal bifurcation rates rise in the cerebral cortex from embryonic day (E)17 and are reduced by postnatal day (P)9. Since callosal neurons populate deep and superficial cortical layers, we compare the axon bifurcation ratio between those neurons by electroporating ex vivo brains at E13 and E15, using eGFP reporter to label the newborn neurons on organotypic slices. Our results suggest that deep layer neurons bifurcate 32% more than superficial ones. To investigate axonal bifurcation in CC dysgenesis, we use BALB/c mice as a spontaneous CC dysgenesis model. BALB/c mice present a typical layer distribution of SATB2 callosal cells, despite the occurrence of callosal anomalies. However, using anterograde DiI tracing, we find that BALB/c mice display increased rates of axonal bifurcations during early and late cortical development in the medial frontal cortex. Midline guidepost cells adjacent to the medial frontal cortex are significant reduced in the CC dysgenesis mouse model. Altogether these data suggest that callosal collateral axonal exuberance is maintained in the absence of midline guidepost signaling and might facilitate aberrant connections in the CC dysgenesis mouse model.
Collapse
Affiliation(s)
- Danielle Rayêe
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Institute of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Pamela Meneses Iack
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil
| | - Raissa R Christoff
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil
| | - Michele R Lourenço
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro - IFRJ, Brazil
| | | | - Jürgen Boltz
- Institute of General Zoology and Animal Physiology, University of Jena, 07743 Jena, Germany
| | - Roberto Lent
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Institute D'Or for Research and Education, Rio de Janeiro, Brazil
| | - Patricia P Garcez
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Wittmann MT, Katada S, Sock E, Kirchner P, Ekici AB, Wegner M, Nakashima K, Lie DC, Reis A. scRNA sequencing uncovers a TCF4-dependent transcription factor network regulating commissure development in mouse. Development 2021; 148:269257. [PMID: 34184026 PMCID: PMC8327186 DOI: 10.1242/dev.196022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 06/15/2021] [Indexed: 01/21/2023]
Abstract
Transcription factor 4 (TCF4) is a crucial regulator of neurodevelopment and has been linked to the pathogenesis of autism, intellectual disability and schizophrenia. As a class I bHLH transcription factor (TF), it is assumed that TCF4 exerts its neurodevelopmental functions through dimerization with proneural class II bHLH TFs. Here, we aim to identify TF partners of TCF4 in the control of interhemispheric connectivity formation. Using a new bioinformatic strategy integrating TF expression levels and regulon activities from single cell RNA-sequencing data, we find evidence that TCF4 interacts with non-bHLH TFs and modulates their transcriptional activity in Satb2+ intercortical projection neurons. Notably, this network comprises regulators linked to the pathogenesis of neurodevelopmental disorders, e.g. FOXG1, SOX11 and BRG1. In support of the functional interaction of TCF4 with non-bHLH TFs, we find that TCF4 and SOX11 biochemically interact and cooperatively control commissure formation in vivo, and regulate the transcription of genes implicated in this process. In addition to identifying new candidate interactors of TCF4 in neurodevelopment, this study illustrates how scRNA-Seq data can be leveraged to predict TF networks in neurodevelopmental processes. Summary: Single-cell RNA sequencing identifies interactions of TCF4 with non-bHLH transcription factors linked to neurodevelopmental and neuropsychiatric disease in the regulation of interhemispheric projection neuron development.
Collapse
Affiliation(s)
- Marie-Theres Wittmann
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany.,Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Sayako Katada
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Elisabeth Sock
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Michael Wegner
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Dieter Chichung Lie
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - André Reis
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
20
|
Cousin MA, Creighton BA, Breau KA, Spillmann RC, Torti E, Dontu S, Tripathi S, Ajit D, Edwards RJ, Afriyie S, Bay JC, Harper KM, Beltran AA, Munoz LJ, Falcon Rodriguez L, Stankewich MC, Person RE, Si Y, Normand EA, Blevins A, May AS, Bier L, Aggarwal V, Mancini GMS, van Slegtenhorst MA, Cremer K, Becker J, Engels H, Aretz S, MacKenzie JJ, Brilstra E, van Gassen KLI, van Jaarsveld RH, Oegema R, Parsons GM, Mark P, Helbig I, McKeown SE, Stratton R, Cogne B, Isidor B, Cacheiro P, Smedley D, Firth HV, Bierhals T, Kloth K, Weiss D, Fairley C, Shieh JT, Kritzer A, Jayakar P, Kurtz-Nelson E, Bernier RA, Wang T, Eichler EE, van de Laar IMBH, McConkie-Rosell A, McDonald MT, Kemppainen J, Lanpher BC, Schultz-Rogers LE, Gunderson LB, Pichurin PN, Yoon G, Zech M, Jech R, Winkelmann J, Beltran AS, Zimmermann MT, Temple B, Moy SS, Klee EW, Tan QKG, Lorenzo DN. Pathogenic SPTBN1 variants cause an autosomal dominant neurodevelopmental syndrome. Nat Genet 2021; 53:1006-1021. [PMID: 34211179 PMCID: PMC8273149 DOI: 10.1038/s41588-021-00886-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 05/14/2021] [Indexed: 12/22/2022]
Abstract
SPTBN1 encodes βII-spectrin, the ubiquitously expressed β-spectrin that forms micrometer-scale networks associated with plasma membranes. Mice deficient in neuronal βII-spectrin have defects in cortical organization, developmental delay and behavioral deficiencies. These phenotypes, while less severe, are observed in haploinsufficient animals, suggesting that individuals carrying heterozygous SPTBN1 variants may also show measurable compromise of neural development and function. Here we identify heterozygous SPTBN1 variants in 29 individuals with developmental, language and motor delays; mild to severe intellectual disability; autistic features; seizures; behavioral and movement abnormalities; hypotonia; and variable dysmorphic facial features. We show that these SPTBN1 variants lead to effects that affect βII-spectrin stability, disrupt binding to key molecular partners, and disturb cytoskeleton organization and dynamics. Our studies define SPTBN1 variants as the genetic basis of a neurodevelopmental syndrome, expand the set of spectrinopathies affecting the brain and underscore the critical role of βII-spectrin in the central nervous system.
Collapse
Affiliation(s)
- Margot A Cousin
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
| | - Blake A Creighton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Keith A Breau
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca C Spillmann
- Department of Pediatrics, Duke University Medical Center, Duke University, Durham, NC, USA
| | | | - Sruthi Dontu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Swarnendu Tripathi
- Bioinformatics Research and Development Laboratory, Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepa Ajit
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Reginald J Edwards
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Simone Afriyie
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Julia C Bay
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kathryn M Harper
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alvaro A Beltran
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Human Pluripotent Stem Cell Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lorena J Munoz
- Human Pluripotent Stem Cell Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Liset Falcon Rodriguez
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Yue Si
- GeneDx, Gaithersburg, MD, USA
| | | | | | - Alison S May
- Department of Neurology, Columbia University, New York, NY, USA
| | - Louise Bier
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Vimla Aggarwal
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
- Laboratory of Personalized Genomic Medicine, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | | | - Kirsten Cremer
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Jessica Becker
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Hartmut Engels
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Stefan Aretz
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | | | - Eva Brilstra
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Koen L I van Gassen
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Renske Oegema
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Paul Mark
- Spectrum Health Medical Genetics, Grand Rapids, MI, USA
| | - Ingo Helbig
- Division of Neurology, Departments of Neurology and Pediatrics, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah E McKeown
- Division of Neurology, Departments of Neurology and Pediatrics, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robert Stratton
- Genetics, Driscoll Children's Hospital, Corpus Christi, TX, USA
| | - Benjamin Cogne
- Service de Génétique Médicale, CHU Nantes, Nantes, France
- Université de Nantes, CNRS, INSERM, L'Institut du Thorax, Nantes, France
| | - Bertrand Isidor
- Service de Génétique Médicale, CHU Nantes, Nantes, France
- Université de Nantes, CNRS, INSERM, L'Institut du Thorax, Nantes, France
| | - Pilar Cacheiro
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Damian Smedley
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Helen V Firth
- Department of Clinical Genetics, Cambridge University Hospitals, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Tatjana Bierhals
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katja Kloth
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Deike Weiss
- Neuropediatrics, Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cecilia Fairley
- Division of Medical Genetics, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Joseph T Shieh
- Division of Medical Genetics, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Amy Kritzer
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | | | - Evangeline Kurtz-Nelson
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Raphael A Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Tianyun Wang
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Ingrid M B H van de Laar
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Allyn McConkie-Rosell
- Department of Pediatrics, Duke University Medical Center, Duke University, Durham, NC, USA
| | - Marie T McDonald
- Department of Pediatrics, Duke University Medical Center, Duke University, Durham, NC, USA
| | - Jennifer Kemppainen
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Brendan C Lanpher
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Laura E Schultz-Rogers
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Lauren B Gunderson
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Pavel N Pichurin
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Grace Yoon
- Divisions of Clinical/Metabolic Genetics and Neurology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
| | - Robert Jech
- Department of Neurology, Charles University, 1st Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Lehrstuhl für Neurogenetik, Technische Universität München, Munich, Germany
- Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
| | - Adriana S Beltran
- Human Pluripotent Stem Cell Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael T Zimmermann
- Bioinformatics Research and Development Laboratory, Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
- Clinical and Translational Sciences Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brenda Temple
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sheryl S Moy
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Queenie K-G Tan
- Department of Pediatrics, Duke University Medical Center, Duke University, Durham, NC, USA
| | - Damaris N Lorenzo
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
21
|
Chen KS, Lynton Z, Lim JWC, Robertson T, Gronostajski RM, Bunt J, Richards LJ. NFIA and NFIB function as tumour suppressors in high-grade glioma in mice. Carcinogenesis 2021; 42:357-368. [PMID: 33346791 DOI: 10.1093/carcin/bgaa139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/05/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022] Open
Abstract
Nuclear factor one (NFI) transcription factors are implicated in both brain development and cancer in mice and humans and play an essential role in glial differentiation. NFI expression is reduced in human astrocytoma samples, particularly those of higher grade, whereas over-expression of NFI protein can induce the differentiation of glioblastoma cells within human tumour xenografts and in glioblastoma cell lines in vitro. These data indicate that NFI proteins may act as tumour suppressors in glioma. To test this hypothesis, we generated complex mouse genetic crosses involving six alleles to target gene deletion of known tumour suppressor genes that induce endogenous high-grade glioma in mice, and overlaid this with loss of function Nfi mutant alleles, Nfia and Nfib, a reporter transgene and an inducible Cre allele. Deletion of Nfi resulted in reduced survival time of the mice, increased tumour load and a more aggressive tumour phenotype than observed in glioma mice with normal expression of NFI. Together, these data indicate that NFI genes represent a credible target for both diagnostic analyses and therapeutic strategies to combat high-grade glioma.
Collapse
Affiliation(s)
- Kok-Siong Chen
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Zorana Lynton
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jonathan W C Lim
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Thomas Robertson
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia.,Anatomical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Jens Bunt
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Linda J Richards
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
22
|
Uehara T, Sanuki R, Ogura Y, Yokoyama A, Yoshida T, Futagawa H, Yoshihashi H, Yamada M, Suzuki H, Takenouchi T, Matsubara K, Hirata H, Kosaki K, Takano‐Shimizu T. Recurrent NFIA K125E substitution represents a loss-of-function allele: Sensitive in vitro and in vivo assays for nontruncating alleles. Am J Med Genet A 2021; 185:2084-2093. [PMID: 33973697 PMCID: PMC8251549 DOI: 10.1002/ajmg.a.62226] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 01/09/2023]
Abstract
Nuclear factor I A (NFIA) is a transcription factor that belongs to the NFI family. Truncating variants or intragenic deletion of the NFIA gene are known to cause the human neurodevelopmental disorder known as NFIA‐related disorder, but no patient heterozygous for a missense mutation has been reported. Here, we document two unrelated patients with typical phenotypic features of the NFIA‐related disorder who shared a missense variant p.Lys125Glu (K125E) in the NFIA gene. Patient 1 was a 6‐year‐old female with global developmental delay, corpus callosum anomaly, macrocephaly, and dysmorphic facial features. Patient 2 was a 14‐month‐old male with corpus callosum anomaly and macrocephaly. By using Drosophila and zebrafish models, we functionally evaluated the effect of the K125E substitution. Ectopic expression of wild‐type human NFIA in Drosophila caused developmental defects such as eye malformation and premature death, while that of human NFIA K125E variant allele did not. nfia‐deficient zebrafish embryos showed defects of midline‐crossing axons in the midbrain/hindbrain boundary. This impairment of commissural neurons was rescued by expression of wild‐type human NFIA, but not by that of mutant variant harboring K125E substitution. In accordance with these in vivo functional analyses, we showed that the K125E mutation impaired the transcriptional regulation of HES1 promoter in cultured cells. Taken together, we concluded that the K125E variant in the NFIA gene is a loss‐of‐function mutation.
Collapse
Affiliation(s)
- Tomoko Uehara
- Center for Medical GeneticsKeio University School of MedicineTokyoJapan
| | - Rikako Sanuki
- Advanced Insect Research Promotion CenterKyoto Institute of TechnologyKyotoJapan
| | - Yurie Ogura
- Department of Chemistry and Biological ScienceCollege of Science and Engineering, Aoyama Gakuin UniversitySagamiharaKanagawaJapan
| | - Atsushi Yokoyama
- Department of PediatricsKyoto University Graduate School of MedicineTokyoJapan
| | - Takeshi Yoshida
- Department of PediatricsKyoto University Graduate School of MedicineTokyoJapan
| | - Hiroshi Futagawa
- Department of GeneticsTokyo Metropolitan Children's Medical CenterTokyoJapan
| | - Hiroshi Yoshihashi
- Department of GeneticsTokyo Metropolitan Children's Medical CenterTokyoJapan
| | - Mamiko Yamada
- Center for Medical GeneticsKeio University School of MedicineTokyoJapan
| | - Hisato Suzuki
- Center for Medical GeneticsKeio University School of MedicineTokyoJapan
| | | | - Kohei Matsubara
- Advanced Insect Research Promotion CenterKyoto Institute of TechnologyKyotoJapan
| | - Hiromi Hirata
- Department of Chemistry and Biological ScienceCollege of Science and Engineering, Aoyama Gakuin UniversitySagamiharaKanagawaJapan
| | - Kenjiro Kosaki
- Center for Medical GeneticsKeio University School of MedicineTokyoJapan
| | | |
Collapse
|
23
|
Yeon GB, Shin WH, Yoo SH, Kim D, Jeon BM, Park WU, Bae Y, Park JY, You S, Na D, Kim DS. NFIB induces functional astrocytes from human pluripotent stem cell-derived neural precursor cells mimicking in vivo astrogliogenesis. J Cell Physiol 2021; 236:7625-7641. [PMID: 33949692 DOI: 10.1002/jcp.30405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 12/18/2022]
Abstract
The ability to generate astrocytes from human pluripotent stem cells (hPSCs) offers a promising cellular model to study the development and physiology of human astrocytes. The extant methods for generating functional astrocytes required long culture periods and there remained much ambiguity on whether such paradigms follow the innate developmental program. In this report, we provided an efficient and rapid method for generating physiologically functional astrocytes from hPSCs. Overexpressing the nuclear factor IB in hPSC-derived neural precursor cells induced a highly enriched astrocyte population in 2 weeks. RNA sequencing and functional analyses demonstrated progressive transcriptomic and physiological changes in the cells, resembling in vivo astrocyte development. Further analyses substantiated previous results and established the MAPK pathway necessary for astrocyte differentiation. Hence, this differentiation paradigm provides a prospective in vitro model for human astrogliogenesis studies and the pathophysiology of neurological diseases concerning astrocytes.
Collapse
Affiliation(s)
- Gyu-Bum Yeon
- Department of Biotechnology, Korea University, Seoul, Korea
| | - Won-Ho Shin
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Korea
| | - Seo Hyun Yoo
- Department of Biotechnology, Korea University, Seoul, Korea
| | - Dongyun Kim
- Department of Biotechnology, Korea University, Seoul, Korea
| | | | - Won-Ung Park
- Department of Biotechnology, Korea University, Seoul, Korea
| | - Yeonju Bae
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Korea
| | - Seungkwon You
- Department of Biotechnology, Korea University, Seoul, Korea
| | - Dokyun Na
- School of Integrative Engineering, Chung-Ang University, Seoul, Korea
| | - Dae-Sung Kim
- Department of Biotechnology, Korea University, Seoul, Korea.,Department of Pediatrics, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
24
|
Morcom L, Edwards TJ, Rider E, Jones-Davis D, Lim JW, Chen KS, Dean RJ, Bunt J, Ye Y, Gobius I, Suárez R, Mandelstam S, Sherr EH, Richards LJ. DRAXIN regulates interhemispheric fissure remodelling to influence the extent of corpus callosum formation. eLife 2021; 10:61618. [PMID: 33945466 PMCID: PMC8137145 DOI: 10.7554/elife.61618] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/01/2021] [Indexed: 12/14/2022] Open
Abstract
Corpus callosum dysgenesis (CCD) is a congenital disorder that incorporates either partial or complete absence of the largest cerebral commissure. Remodelling of the interhemispheric fissure (IHF) provides a substrate for callosal axons to cross between hemispheres, and its failure is the main cause of complete CCD. However, it is unclear whether defects in this process could give rise to the heterogeneity of expressivity and phenotypes seen in human cases of CCD. We identify incomplete IHF remodelling as the key structural correlate for the range of callosal abnormalities in inbred and outcrossed BTBR mouse strains, as well as in humans with partial CCD. We identify an eight base-pair deletion in Draxin and misregulated astroglial and leptomeningeal proliferation as genetic and cellular factors for variable IHF remodelling and CCD in BTBR strains. These findings support a model where genetic events determine corpus callosum structure by influencing leptomeningeal-astroglial interactions at the IHF.
Collapse
Affiliation(s)
- Laura Morcom
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Timothy J Edwards
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia.,Faculty of Medicine, Brisbane, Australia
| | - Eric Rider
- Departments of Neurology and Pediatrics, Institute of Human Genetics and Weill Institute of Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Dorothy Jones-Davis
- Departments of Neurology and Pediatrics, Institute of Human Genetics and Weill Institute of Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Jonathan Wc Lim
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Kok-Siong Chen
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Ryan J Dean
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Jens Bunt
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Yunan Ye
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Ilan Gobius
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Rodrigo Suárez
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Simone Mandelstam
- Department of Radiology, University of Melbourne, Royal Children's Hospital, Parkville, Australia
| | - Elliott H Sherr
- Departments of Neurology and Pediatrics, Institute of Human Genetics and Weill Institute of Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Linda J Richards
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia.,School of Biomedical Sciences, Brisbane, Australia
| |
Collapse
|
25
|
Fenlon LR, Suarez R, Lynton Z, Richards LJ. The evolution, formation and connectivity of the anterior commissure. Semin Cell Dev Biol 2021; 118:50-59. [PMID: 33958283 DOI: 10.1016/j.semcdb.2021.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 10/21/2022]
Abstract
The anterior commissure is the most ancient of the forebrain interhemispheric connections among all vertebrates. Indeed, it is the predominant pallial commissure in all non-eutherian vertebrates, universally subserving basic functions related to olfaction and survival. A key feature of the anterior commissure is its ability to convey connections from diverse brain areas, such as most of the neocortex in non-eutherian mammals, thereby mediating the bilateral integration of diverse functions. Shared developmental mechanisms between the anterior commissure and more evolutionarily recent commissures, such as the corpus callosum in eutherians, have led to the hypothesis that the former may have been a precursor for additional expansion of commissural circuits. However, differences between the formation of the anterior commissure and other telencephalic commissures suggest that independent developmental mechanisms underlie the emergence of these connections in extant species. Here, we review the developmental mechanisms and connectivity of the anterior commissure across evolutionarily distant species, and highlight its potential functional importance in humans, both in the course of normal neurodevelopment, and as a site of plastic axonal rerouting in the absence or damage of other connections.
Collapse
Affiliation(s)
- Laura R Fenlon
- The University of Queensland, The Queensland Brain Institute, Brisbane, Australia.
| | - Rodrigo Suarez
- The University of Queensland, The Queensland Brain Institute, Brisbane, Australia
| | - Zorana Lynton
- The University of Queensland, The Queensland Brain Institute, Brisbane, Australia; The Faculty of Medicine, Brisbane, Australia
| | - Linda J Richards
- The University of Queensland, The Queensland Brain Institute, Brisbane, Australia; The School of Biomedical Sciences, Brisbane, Australia.
| |
Collapse
|
26
|
Ezan J, Moreau MM, Mamo TM, Shimbo M, Decroo M, Richter M, Peyroutou R, Rachel R, Tissir F, de Anda FC, Sans N, Montcouquiol M. Early loss of Scribble affects cortical development, interhemispheric connectivity and psychomotor activity. Sci Rep 2021; 11:9106. [PMID: 33907211 PMCID: PMC8079449 DOI: 10.1038/s41598-021-88147-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 04/01/2021] [Indexed: 12/03/2022] Open
Abstract
Neurodevelopmental disorders arise from combined defects in processes including cell proliferation, differentiation, migration and commissure formation. The evolutionarily conserved tumor-suppressor protein Scribble (Scrib) serves as a nexus to transduce signals for the establishment of apicobasal and planar cell polarity during these processes. Human SCRIB gene mutations are associated with neural tube defects and this gene is located in the minimal critical region deleted in the rare Verheij syndrome. In this study, we generated brain-specific conditional cKO mouse mutants and assessed the impact of the Scrib deletion on brain morphogenesis and behavior. We showed that embryonic deletion of Scrib in the telencephalon leads to cortical thickness reduction (microcephaly) and partial corpus callosum and hippocampal commissure agenesis. We correlated these phenotypes with a disruption in various developmental mechanisms of corticogenesis including neurogenesis, neuronal migration and axonal connectivity. Finally, we show that Scrib cKO mice have psychomotor deficits such as locomotor activity impairment and memory alterations. Altogether, our results show that Scrib is essential for early brain development due to its role in several developmental cellular mechanisms that could underlie some of the deficits observed in complex neurodevelopmental pathologies.
Collapse
Affiliation(s)
- Jerome Ezan
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France.
| | - Maité M Moreau
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Tamrat M Mamo
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Miki Shimbo
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Maureen Decroo
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Melanie Richter
- Germany Center for Molecular Neurobiology Hamburg (ZMNH), Research Group Neuronal Development, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronan Peyroutou
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Rivka Rachel
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, NIH, Bethesda, MD, 20892, USA
| | - Fadel Tissir
- Developmental Neurobiology Group, Institute of Neuroscience, University of Louvain, Avenue Mounier 73, Box B1.73.16, 1200, Brussels, Belgium
| | - Froylan Calderon de Anda
- Germany Center for Molecular Neurobiology Hamburg (ZMNH), Research Group Neuronal Development, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Sans
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France
| | - Mireille Montcouquiol
- Université de Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077, Bordeaux, France.
| |
Collapse
|
27
|
Morcom L, Gobius I, Marsh APL, Suárez R, Lim JWC, Bridges C, Ye Y, Fenlon LR, Zagar Y, Douglass AM, Donahoo ALS, Fothergill T, Shaikh S, Kozulin P, Edwards TJ, Cooper HM, IRC5 Consortium, Sherr EH, Chédotal A, Leventer RJ, Lockhart PJ, Richards LJ. DCC regulates astroglial development essential for telencephalic morphogenesis and corpus callosum formation. eLife 2021; 10:e61769. [PMID: 33871356 PMCID: PMC8116049 DOI: 10.7554/elife.61769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 04/18/2021] [Indexed: 02/04/2023] Open
Abstract
The forebrain hemispheres are predominantly separated during embryogenesis by the interhemispheric fissure (IHF). Radial astroglia remodel the IHF to form a continuous substrate between the hemispheres for midline crossing of the corpus callosum (CC) and hippocampal commissure (HC). Deleted in colorectal carcinoma (DCC) and netrin 1 (NTN1) are molecules that have an evolutionarily conserved function in commissural axon guidance. The CC and HC are absent in Dcc and Ntn1 knockout mice, while other commissures are only partially affected, suggesting an additional aetiology in forebrain commissure formation. Here, we find that these molecules play a critical role in regulating astroglial development and IHF remodelling during CC and HC formation. Human subjects with DCC mutations display disrupted IHF remodelling associated with CC and HC malformations. Thus, axon guidance molecules such as DCC and NTN1 first regulate the formation of a midline substrate for dorsal commissures prior to their role in regulating axonal growth and guidance across it.
Collapse
Affiliation(s)
- Laura Morcom
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Ilan Gobius
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Ashley PL Marsh
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children’s Research Institute, Royal Children’s HospitalParkvilleAustralia
- Department of Paediatrics, University of MelbourneParkvilleAustralia
| | - Rodrigo Suárez
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Jonathan WC Lim
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Caitlin Bridges
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Yunan Ye
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Laura R Fenlon
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la VisionParisFrance
| | - Amelia M Douglass
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | | | - Thomas Fothergill
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Samreen Shaikh
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Peter Kozulin
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - Timothy J Edwards
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
- The University of Queensland, Faculty of MedicineBrisbaneAustralia
| | - Helen M Cooper
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
| | - IRC5 Consortium
- Members and Affiliates of the International Research Consortium for the Corpus Callosum and Cerebral Connectivity (IRC5)Los AngelesUnited States
| | - Elliott H Sherr
- Departments of Neurology and Pediatrics, Institute of Human Genetics and Weill Institute of Neurosciences, University of California, San FranciscoSan FranciscoUnited States
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la VisionParisFrance
| | - Richard J Leventer
- Department of Paediatrics, University of MelbourneParkvilleAustralia
- Neuroscience Research Group, Murdoch Children’s Research InstituteParkvilleAustralia
- Department of Neurology, University of Melbourne, Royal Children’s HospitalParkvilleAustralia
| | - Paul J Lockhart
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children’s Research Institute, Royal Children’s HospitalParkvilleAustralia
- Department of Paediatrics, University of MelbourneParkvilleAustralia
| | - Linda J Richards
- The University of Queensland, Queensland Brain InstituteBrisbaneAustralia
- The University of Queensland, School of Biomedical SciencesBrisbaneAustralia
| |
Collapse
|
28
|
Barker MS, Knight JL, Dean RJ, Mandelstam S, Richards LJ, Robinson GA. Verbal Adynamia and Conceptualization in Partial Rhombencephalosynapsis and Corpus Callosum Dysgenesis. Cogn Behav Neurol 2021; 34:38-52. [PMID: 33652468 DOI: 10.1097/wnn.0000000000000261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/02/2020] [Indexed: 11/26/2022]
Abstract
Verbal adynamia is characterized by markedly reduced spontaneous speech that is not attributable to a core language deficit such as impaired naming, reading, repetition, or comprehension. In some cases, verbal adynamia is severe enough to be considered dynamic aphasia. We report the case of a 40-year-old, left-handed, male native English speaker who presented with partial rhombencephalosynapsis, corpus callosum dysgenesis, and a language profile that is consistent with verbal adynamia, or subclinical dynamic aphasia, possibly underpinned by difficulties selecting and generating ideas for expression. This case is only the second investigation of dynamic aphasia in an individual with a congenital brain malformation. It is also the first detailed neuropsychological report of an adult with partial rhombencephalosynapsis and corpus callosum dysgenesis, and the only known case of superior intellectual abilities in this context.
Collapse
Affiliation(s)
- Megan S Barker
- Neuropsychology Research Unit, School of Psychology, The University of Queensland, St Lucia, Brisbane, Australia
- Taub Institute, Columbia University Medical Center, New York, New York
| | - Jacquelyn L Knight
- Neuropsychology Research Unit, School of Psychology, The University of Queensland, St Lucia, Brisbane, Australia
| | - Ryan J Dean
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Australia
| | - Simone Mandelstam
- Department of Radiology, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
| | - Linda J Richards
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Brisbane, Australia
| | - Gail A Robinson
- Neuropsychology Research Unit, School of Psychology, The University of Queensland, St Lucia, Brisbane, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Australia
| |
Collapse
|
29
|
Kim BH, Nho K, Lee JM. Genome-wide association study identifies susceptibility loci of brain atrophy to NFIA and ST18 in Alzheimer's disease. Neurobiol Aging 2021; 102:200.e1-200.e11. [PMID: 33640202 DOI: 10.1016/j.neurobiolaging.2021.01.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/08/2021] [Accepted: 01/25/2021] [Indexed: 02/04/2023]
Abstract
To identify genetic variants influencing cortical atrophy in Alzheimer's disease (AD), we performed genome-wide association studies (GWAS) of mean cortical thicknesses in 17 AD-related brain. In this study, we used neuroimaging and genetic data of 919 participants from the Alzheimer's Disease Neuroimaging Initiative cohort, which include 268 cognitively normal controls, 488 mild cognitive impairment, 163 AD individuals. We performed GWAS with 3,041,429 single nucleotide polymorphisms (SNPs) for cortical thickness. The results of GWAS indicated that rs10109716 in ST18 (ST18 C2H2C-type zinc finger transcription factor) and rs661526 in NFIA (nuclear factor I A) genes are significantly associated with mean cortical thicknesses of the left inferior frontal gyrus and left parahippocampal gyrus, respectively. The rs661526 regulates the expression levels of NFIA in the substantia nigra and frontal cortex and rs10109716 regulates the expression levels of ST18 in the thalamus. These results suggest a crucial role of identified genes for cortical atrophy and could provide further insights into the genetic basis of AD.
Collapse
Affiliation(s)
- Bo-Hyun Kim
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Jong-Min Lee
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea.
| | | |
Collapse
|
30
|
De León Reyes NS, Bragg-Gonzalo L, Nieto M. Development and plasticity of the corpus callosum. Development 2020; 147:147/18/dev189738. [PMID: 32988974 DOI: 10.1242/dev.189738] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The corpus callosum (CC) connects the cerebral hemispheres and is the major mammalian commissural tract. It facilitates bilateral sensory integration and higher cognitive functions, and is often affected in neurodevelopmental diseases. Here, we review the mechanisms that contribute to the development of CC circuits in animal models and humans. These species comparisons reveal several commonalities. First, there is an early period of massive axonal projection. Second, there is a postnatal temporal window, varying between species, in which early callosal projections are selectively refined. Third, sensory-derived activity influences axonal refinement. We also discuss how defects in CC formation can lead to mild or severe CC congenital malformations.
Collapse
Affiliation(s)
- Noelia S De León Reyes
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Lorena Bragg-Gonzalo
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Marta Nieto
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
31
|
Barrus K, Rego S, Yip T, Martin PM, Glen OA, Van Ziffle J, Slavotinek AM. The expanding spectrum of NFIB-associated phenotypes in a diverse patient population-A report of two new patients. Am J Med Genet A 2020; 182:2959-2963. [PMID: 32902921 DOI: 10.1002/ajmg.a.61852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/25/2020] [Accepted: 08/04/2020] [Indexed: 11/07/2022]
Abstract
NFIB (Nuclear Factor I B) haploinsufficiency has recently been identified as a cause of intellectual disability and macrocephaly. Here we describe two patients with pathogenic variants in NFIB. The first is a 6-year-old Latino male with developmental delays, mild hypotonia, facial anomalies, and brain magnetic resonance imaging findings comprising mild thinning of the corpus callosum, with more marked thinning of the splenium and blunting of the rostrum and cavum septum pellucidum. Exome sequencing identified a previously described de novo variant in NFIB, c.265C>T, predicting p.Arg89Ter. The second is a 5-year-old Latino male with developmental delays, hypotonia, dysmorphic features, a preauricular tag and pit, a small ventricular septal defect, and brain magnetic resonance imaging findings including a dysmorphic corpus callosum and a small posterior fossa. A single nucleotide polymorphism microarray identified a 92 kb interstitial deletion at 9p23 including several exons of NFIB and no other known genes. Our two patients add to the knowledge of this rare condition through our addition of new brain MRI findings and dysmorphic features. Additionally, these are the first known Latino patients to be described with NFIB haploinsufficiency, expanding our understanding of the associated facial features in diverse populations. Further data are needed to determine genotype-phenotype relationships for NFIB.
Collapse
Affiliation(s)
- Kathleen Barrus
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Shannon Rego
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Tiffany Yip
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Pierre-Marie Martin
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Orit A Glen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Jessica Van Ziffle
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Anne M Slavotinek
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA.,Division of Medical Genetics, Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
32
|
Shakes P, Cashin A, Hurley J. Scoping Review of the Prenatal Diagnosis of Agenesis of the Corpus Callosum. J Obstet Gynecol Neonatal Nurs 2020; 49:423-436. [PMID: 32687791 DOI: 10.1016/j.jogn.2020.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2020] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To map and summarize the literature related to the prenatal diagnosis of agenesis of the corpus callosum (ACC) to inform nursing practice. DATA SOURCES We searched MEDLINE, CINAHL, PyscINFO, and Academic Search Complete with the use of strings of curated terms to cover the broad ACC nomenclature. Documents were published in English between 2009 and June 1, 2020. We also hand searched the reference lists of included documents. STUDY SELECTION We screened 582 abstracts and retrieved the full texts of primary research articles, reviews, discussion papers, and peer-reviewed book chapters if the abstracts specifically mentioned ACC and the prenatal period. We excluded case reports, conference and poster abstracts, papers on broader anomalies, and animal studies. We reviewed 84 full-text documents and identified 61 for inclusion. DATA EXTRACTION We charted the data through an iterative process under headings for location, article type, study design, participant age, ACC type, recruitment, method, tools/assessments, results, key recommendations, gestational age at diagnosis, termination of pregnancy rate, the definition of isolated ACC, and our notes of critique of the document. DATA SYNTHESIS We constructed a narrative synthesis from thematically arranged data. In the included documents, ACC was diagnosed between 17 and 38 weeks gestation and was frequently described as heterogeneous because of different causes, presentations, and outcomes. Whether the ACC was isolated as the only anomaly or present with other anomalies was considered the key factor for prenatal counseling. However, the definition of isolated ACC was inconsistent. CONCLUSION The inconsistent nomenclature and definitions of an isolated presentation of ACC increase the ambiguity in the prenatal diagnosis and must be considered when the outcome and diagnostic efficacy studies are interpreted. There is an absence of research on parents' experiences of prenatal diagnoses of ACC to inform holistic nursing interventions and the provision of psychosocial support.
Collapse
|
33
|
Mesman S, Bakker R, Smidt MP. Tcf4 is required for correct brain development during embryogenesis. Mol Cell Neurosci 2020; 106:103502. [PMID: 32474139 DOI: 10.1016/j.mcn.2020.103502] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/28/2020] [Accepted: 05/19/2020] [Indexed: 01/02/2023] Open
Abstract
Tcf4 has been linked to autism, schizophrenia, and Pitt-Hopkins Syndrome (PTHS) in humans, suggesting a role for Tcf4 in brain development and importantly cortical development. However, the mechanisms behind its role in disease and brain development are still elusive. We provide evidence that Tcf4 has a critical function in the differentiation of cortical regions, corpus callosum and anterior commissure formation, and development of the hippocampus during murine embryonic development. In the present study, we show that Tcf4 is expressed throughout the developing brain at the peak of neurogenesis. Deletion of Tcf4 results in mis-specification of the cortical neurons, malformation of the corpus callosum and anterior commissure, and hypoplasia of the hippocampus. Furthermore, the Tcf4 mutant shows an absence of midline remodeling, underlined by the loss of GFAP-expressing midline glia in the indusium griseum and callosal wedge and midline zipper glia in the telencephalic midline. RNA-sequencing on E14.5 cortex material shows that Tcf4 functions as a transcriptional activator and loss of Tcf4 results in downregulation of genes linked to neurogenesis and neuronal maturation. Furthermore, many genes that are differentially expressed after Tcf4 ablation are linked to other neurodevelopmental disorders. Taken together, we show that correct brain development and neuronal differentiation are severely affected in Tcf4 mutants, phenocopying morphological brain defects detected in PTHS patients. The presented data identifies new leads to understand the mechanisms behind brain and specifically cortical development and can provide novel insights in developmental mechanisms underlying human brain defects.
Collapse
Affiliation(s)
- Simone Mesman
- Swammerdam Institute for Life Sciences, FNWI University of Amsterdam, Science Park 904, 1098XH Amsterdam, the Netherlands
| | - Reinier Bakker
- Swammerdam Institute for Life Sciences, FNWI University of Amsterdam, Science Park 904, 1098XH Amsterdam, the Netherlands
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, FNWI University of Amsterdam, Science Park 904, 1098XH Amsterdam, the Netherlands.
| |
Collapse
|
34
|
Charvet CJ, Das A, Song JW, Tindal-Burgess DJ, Kabaria P, Dai G, Kane T, Takahashi E. High Angular Resolution Diffusion MRI Reveals Conserved and Deviant Programs in the Paths that Guide Human Cortical Circuitry. Cereb Cortex 2020; 30:1447-1464. [PMID: 31667494 PMCID: PMC7132938 DOI: 10.1093/cercor/bhz178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/13/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
Diffusion magnetic resonance (MR) tractography represents a novel opportunity to investigate conserved and deviant developmental programs between humans and other species such as mice. To that end, we acquired high angular resolution diffusion MR scans of mice [embryonic day (E) 10.5 to postnatal week 4] and human brains [gestational week (GW) 17-30] at successive stages of fetal development to investigate potential evolutionary changes in radial organization and emerging pathways between humans and mice. We compare radial glial development as well as commissural development (e.g., corpus callosum), primarily because our findings can be integrated with previous work. We also compare corpus callosal growth trajectories across primates (i.e., humans and rhesus macaques) and rodents (i.e., mice). One major finding is that the developing cortex of humans is predominated by pathways likely associated with a radial glial organization at GW 17-20, which is not as evident in age-matched mice (E 16.5, 17.5). Another finding is that, early in development, the corpus callosum follows a similar developmental timetable in primates (i.e., macaques and humans) as in mice. However, the corpus callosum grows for an extended period of time in primates compared with rodents. Taken together, these findings highlight deviant developmental programs underlying the emergence of cortical pathways in the human brain.
Collapse
Affiliation(s)
| | - Avilash Das
- Medical Sciences in the College of Arts and Sciences, Boston University, Boston, MA 02215, USA
- Division of Newborn Medicine, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02215, USA
- Fetal-Neonatal Brain Imaging and Developmental Science Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Jae W Song
- Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Priya Kabaria
- Department of Behavioral Neuroscience, Northeastern University, Boston, MA 02115, USA
| | - Guangping Dai
- Science Center, Wellesley College, Wellesley, MA 02481, USA
| | - Tara Kane
- Department of Behavioral Neuroscience, Northeastern University, Boston, MA 02115, USA
| | - Emi Takahashi
- Division of Newborn Medicine, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02215, USA
- Fetal-Neonatal Brain Imaging and Developmental Science Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02215, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
35
|
Putoux A, Baas D, Paschaki M, Morlé L, Maire C, Attié-Bitach T, Thomas S, Durand B. Altered GLI3 and FGF8 signaling underlies acrocallosal syndrome phenotypes in Kif7 depleted mice. Hum Mol Genet 2020; 28:877-887. [PMID: 30445565 DOI: 10.1093/hmg/ddy392] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 11/14/2022] Open
Abstract
Acrocallosal syndrome (ACLS) is a rare genetic disorder characterized by agenesis or hypoplasia of corpus callosum (CC), polydactyly, craniofacial dysmorphism and severe intellectual deficiency. We previously identified KIF7, a key ciliary component of the Sonic hedgehog (SHH) pathway, as being a causative gene for this syndrome, thus including ACLS in the group of ciliopathies. In both humans and mice, KIF7 depletion leads to abnormal GLI3 processing and over-activation of SHH target genes. To understand the pathological mechanisms involved in CC defects in this syndrome, we took advantage of a previously described Kif7-/- mouse model to demonstrate that in addition to polydactyly and neural tube closure defects, these mice present CC agenesis with characteristic Probst bundles, thus recapitulating major ACLS features. We show that CC agenesis in these mice is associated with specific patterning defects of the cortical septum boundary leading to altered distribution of guidepost cells required to guide the callosal axons through the midline. Furthermore, by crossing Kif7-/- mice with Gli3Δ699 mice exclusively producing the repressive isoform of GLI3 (GLI3R), we demonstrate that decreased GLI3R signaling is fully responsible for the ACLS features in these mice, as all phenotypes are rescued by increasing GLI3R activity. Moreover, we show that increased FGF8 signaling is responsible in part for CC defects associated to KIF7 depletion, as modulating FGF8 signaling rescued CC formation anteriorly in Kif7-/- mice. Taken together our data demonstrate that ACLS features rely on defective GLI3R and FGF8 signaling.
Collapse
Affiliation(s)
- Audrey Putoux
- Centre de Recherche en Neurosciences de Lyon, Équipe GENDEV, INSERM U1028, CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France.,Service de Génétique et Centre de Référence des Anomalies du Développement de la Région Auvergne-Rhône-Alpes, CHU de Lyon, France
| | - Dominique Baas
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U-1217, Lyon, France
| | - Marie Paschaki
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U-1217, Lyon, France
| | - Laurette Morlé
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U-1217, Lyon, France
| | - Charline Maire
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U-1217, Lyon, France
| | - Tania Attié-Bitach
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR1163, Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Department of Histology-Embryology and Cytogenetics, Necker Hospital, AP-HP, Paris, France
| | - Sophie Thomas
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR1163, Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Bénédicte Durand
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U-1217, Lyon, France
| |
Collapse
|
36
|
Chen KS, Bridges CR, Lynton Z, Lim JWC, Stringer BW, Rajagopal R, Wong KT, Ganesan D, Ariffin H, Day BW, Richards LJ, Bunt J. Transcription factors NFIA and NFIB induce cellular differentiation in high-grade astrocytoma. J Neurooncol 2019; 146:41-53. [PMID: 31760595 DOI: 10.1007/s11060-019-03352-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/12/2019] [Accepted: 11/16/2019] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Malignant astrocytomas are composed of heterogeneous cell populations. Compared to grade IV glioblastoma, low-grade astrocytomas have more differentiated cells and are associated with a better prognosis. Therefore, inducing cellular differentiation to alter the behaviour of high-grade astrocytomas may serve as a therapeutic strategy. The nuclear factor one (NFI) transcription factors are essential for normal astrocytic differentiation. Here, we investigate whether family members NFIA and NFIB act as effectors of cellular differentiation in glioblastoma. METHODS We analysed expression of NFIA and NFIB in mRNA expression data of high-grade astrocytoma and with immunofluorescence co-staining. Furthermore, we induced NFI expression in patient-derived subcutaneous glioblastoma xenografts via in vivo electroporation. RESULTS The expression of NFIA and NFIB is reduced in glioblastoma as compared to lower grade astrocytomas. At a cellular level, their expression is associated with differentiated and mature astrocyte-like tumour cells. In vivo analyses consistently demonstrate that expression of either NFIA or NFIB is sufficient to promote tumour cell differentiation in glioblastoma xenografts. CONCLUSION Our findings indicate that both NFIA and NFIB may have an endogenous pro-differentiative function in astrocytomas, similar to their role in normal astrocyte differentiation. Overall, our study establishes a basis for further investigation of targeting NFI-mediated differentiation as a potential differentiation therapy.
Collapse
Affiliation(s)
- Kok-Siong Chen
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Caitlin R Bridges
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zorana Lynton
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
- The Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jonathan W C Lim
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Brett W Stringer
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Revathi Rajagopal
- Department of Paediatrics, University of Malaya, 59100, Kuala Lumpur, Malaysia
| | - Kum-Thong Wong
- Department of Pathology, University of Malaya, 59100, Kuala Lumpur, Malaysia
| | - Dharmendra Ganesan
- Division of Neurosurgery, University of Malaya Medical Centre, 59100, Kuala Lumpur, Malaysia
| | - Hany Ariffin
- Department of Paediatrics, University of Malaya, 59100, Kuala Lumpur, Malaysia
| | - Bryan W Day
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Linda J Richards
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Biomedical Sciences, The Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia.
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Rd Brisbane, Brisbane, QLD, 4072, Australia.
| | - Jens Bunt
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Rd Brisbane, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
37
|
Roig-Puiggros S, Vigouroux RJ, Beckman D, Bocai NI, Chiou B, Davimes J, Gomez G, Grassi S, Hoque A, Karikari TK, Kiffer F, Lopez M, Lunghi G, Mazengenya P, Meier S, Olguín-Albuerne M, Oliveira MM, Paraíso-Luna J, Pradhan J, Radiske A, Ramos-Hryb AB, Ribeiro MC, Schellino R, Selles MC, Singh S, Theotokis P, Chédotal A. Construction and reconstruction of brain circuits: normal and pathological axon guidance. J Neurochem 2019; 153:10-32. [PMID: 31630412 DOI: 10.1111/jnc.14900] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
Perception of our environment entirely depends on the close interaction between the central and peripheral nervous system. In order to communicate each other, both systems must develop in parallel and in coordination. During development, axonal projections from the CNS as well as the PNS must extend over large distances to reach their appropriate target cells. To do so, they read and follow a series of axon guidance molecules. Interestingly, while these molecules play critical roles in guiding developing axons, they have also been shown to be critical in other major neurodevelopmental processes, such as the migration of cortical progenitors. Currently, a major hurdle for brain repair after injury or neurodegeneration is the absence of axonal regeneration in the mammalian CNS. By contrasts, PNS axons can regenerate. Many hypotheses have been put forward to explain this paradox but recent studies suggest that hacking neurodevelopmental mechanisms may be the key to promote CNS regeneration. Here we provide a seminar report written by trainees attending the second Flagship school held in Alpbach, Austria in September 2018 organized by the International Society for Neurochemistry (ISN) together with the Journal of Neurochemistry (JCN). This advanced school has brought together leaders in the fields of neurodevelopment and regeneration in order to discuss major keystones and future challenges in these respective fields.
Collapse
Affiliation(s)
| | - Robin J Vigouroux
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Danielle Beckman
- California National Primate Research Center, UC Davis, Davis, California, USA
| | - Nadia I Bocai
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Brian Chiou
- Department of Pediatrics, University of California - San Francisco, San Francisco, California, USA
| | - Joshua Davimes
- Faculty of Health Sciences School of Anatomical Sciences, University of the Witwatersrand, Parktown Johannesburg, South Africa
| | - Gimena Gomez
- Laboratorio de Parkinson Experimental, Instituto de Investigaciones Farmacológicas (ININFA-CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Ashfaqul Hoque
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,School of Life Sciences, University of Warwick, Coventry, UK.,Midlands Integrative Biosciences Training Partnership, University of Warwick, Coventry, UK
| | - Frederico Kiffer
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Mary Lopez
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicin, University of Milano, Segrate, Italy
| | - Pedzisai Mazengenya
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sonja Meier
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mauricio M Oliveira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juan Paraíso-Luna
- Ramón y Cajal Institute of Health Research (IRYCIS), Department of Biochemistry and Molecular Biology and University Research Institute in Neurochemistry (IUIN), Complutense University, Madrid, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jonu Pradhan
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Andressa Radiske
- Memory Research Laboratory, Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Ana Belén Ramos-Hryb
- Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina.,Grupo de Neurociencia de Sistemas, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Mayara C Ribeiro
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York, USA
| | - Roberta Schellino
- Neuroscience Department "Rita Levi-Montalcini" and Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Torino, Italy
| | - Maria Clara Selles
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Shripriya Singh
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Paschalis Theotokis
- Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Macedonia, Greece
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
38
|
Zenker M, Bunt J, Schanze I, Schanze D, Piper M, Priolo M, Gerkes EH, Gronostajski RM, Richards LJ, Vogt J, Wessels MW, Hennekam RC. Variants in nuclear factor I genes influence growth and development. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2019; 181:611-626. [DOI: 10.1002/ajmg.c.31747] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/26/2019] [Accepted: 10/09/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Martin Zenker
- Institute of Human GeneticsUniversity Hospital, Otto‐von‐Guericke‐University Magdeburg Germany
| | - Jens Bunt
- Queensland Brain InstituteThe University of Queensland Brisbane Queensland Australia
| | - Ina Schanze
- Institute of Human GeneticsUniversity Hospital, Otto‐von‐Guericke‐University Magdeburg Germany
| | - Denny Schanze
- Institute of Human GeneticsUniversity Hospital, Otto‐von‐Guericke‐University Magdeburg Germany
| | - Michael Piper
- Queensland Brain InstituteThe University of Queensland Brisbane Queensland Australia
- School of Biomedical SciencesThe University of Queensland Brisbane Queensland Australia
| | - Manuela Priolo
- Operative Unit of Medical GeneticsGreat Metropolitan Hospital Bianchi‐Melacrino‐Morelli Reggio Calabria Italy
| | - Erica H. Gerkes
- Department of Genetics, University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Richard M. Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life SciencesState University of New York Buffalo NY
| | - Linda J. Richards
- Queensland Brain InstituteThe University of Queensland Brisbane Queensland Australia
- School of Biomedical SciencesThe University of Queensland Brisbane Queensland Australia
| | - Julie Vogt
- West Midlands Regional Clinical Genetics Service and Birmingham Health PartnersWomen's and Children's Hospitals NHS Foundation Trust Birmingham UK
| | - Marja W. Wessels
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center Rotterdam Rotterdam The Netherlands
| | - Raoul C. Hennekam
- Department of PediatricsUniversity of Amsterdam Amsterdam The Netherlands
| |
Collapse
|
39
|
Durán-Carabali LE, Sanches EF, Odorcyk FK, Nicola F, Mestriner RG, Reichert L, Aristimunha D, Pagnussat AS, Netto CA. Tissue Injury and Astrocytic Reaction, But Not Cognitive Deficits, Are Dependent on Hypoxia Duration in Very Immature Rats Undergoing Neonatal Hypoxia-Ischemia. Neurochem Res 2019; 44:2631-2642. [PMID: 31564017 DOI: 10.1007/s11064-019-02884-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/11/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022]
Abstract
Preterm birth and hypoxia-ischemia (HI) are major causes of neonatal death and neurological disabilities in newborns. The widely used preclinical HI model combines carotid occlusion with hypoxia exposure; however, the relationship between different hypoxia exposure periods with brain tissue loss, astrocyte reactivity and behavioral impairments following HI is lacking. Present study evaluated HI-induced behavioral and morphological consequences in rats exposed to different periods of hypoxia at postnatal day 3. Wistar rats of both sexes were assigned into four groups: control group, HI-120 min, HI-180 min and HI-210 min. Neurodevelopmental reflexes, exploratory abilities and cognitive function were assessed. At adulthood, tissue damage and reactive astrogliosis were measured. Animals exposed to HI-180 and HI-210 min had delayed neurodevelopmental reflexes compared to control group. Histological assessment showed tissue loss that was restricted to the ipsilateral hemisphere in lower periods of hypoxia exposure (120 and 180 min) but affected both hemispheres when 210 min was used. Reactive astrogliosis was increased only after 210 min of hypoxia. Interestingly, cognitive deficits were induced regardless the duration of hypoxia and there were correlations between behavioral parameters and cortex, hippocampus and corpus callosum volumes. These results show the duration of hypoxia has a close relationship with astrocytic response and tissue damage progression. Furthermore, the long-lasting cognitive memory deficit and its association with brain structures beyond the hippocampus suggests that complex anatomical changes should be involved in functional alterations taking place as hypoxia duration is increased, even when the cognitive impairment limit is achieved.
Collapse
Affiliation(s)
- L E Durán-Carabali
- Post-graduation Program of Physiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2600, anexo, Porto Alegre, RS, CEP 90035-003, Brazil.
| | - E F Sanches
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - F K Odorcyk
- Post-graduation Program of Physiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2600, anexo, Porto Alegre, RS, CEP 90035-003, Brazil
| | - F Nicola
- Post-graduation Program of Neuroscience, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - R G Mestriner
- Neurorehabilitation and Neural Repair Research Group, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - L Reichert
- Neurorehabilitation and Neural Repair Research Group, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - D Aristimunha
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - A S Pagnussat
- Rehabilitation Sciences Graduate Program, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - C A Netto
- Post-graduation Program of Physiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2600, anexo, Porto Alegre, RS, CEP 90035-003, Brazil.,Post-graduation Program of Neuroscience, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| |
Collapse
|
40
|
Calloni SF, Caschera L, Triulzi FM. Disorders of Ventral Induction/Spectrum of Holoprosencephaly. Neuroimaging Clin N Am 2019; 29:411-421. [DOI: 10.1016/j.nic.2019.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
41
|
Filatova A, Rey LK, Lechler MB, Schaper J, Hempel M, Posmyk R, Szczaluba K, Santen GWE, Wieczorek D, Nuber UA. Mutations in SMARCB1 and in other Coffin-Siris syndrome genes lead to various brain midline defects. Nat Commun 2019; 10:2966. [PMID: 31273213 PMCID: PMC6609698 DOI: 10.1038/s41467-019-10849-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 06/05/2019] [Indexed: 01/09/2023] Open
Abstract
Mutations in genes encoding components of BAF (BRG1/BRM-associated factor) chromatin remodeling complexes cause neurodevelopmental disorders and tumors. The mechanisms leading to the development of these two disease entities alone or in combination remain unclear. We generated mice with a heterozygous nervous system-specific partial loss-of-function mutation in a BAF core component gene, Smarcb1. These Smarcb1 mutant mice show various brain midline abnormalities that are also found in individuals with Coffin–Siris syndrome (CSS) caused by SMARCB1, SMARCE1, and ARID1B mutations and in SMARCB1-related intellectual disability (ID) with choroid plexus hyperplasia (CPH). Analyses of the Smarcb1 mutant animals indicate that one prominent midline abnormality, corpus callosum agenesis, is due to midline glia aberrations. Our results establish a novel role of Smarcb1 in the development of the brain midline and have important clinical implications for BAF complex-related ID/neurodevelopmental disorders. Why and how mutations in genes encoding BAF complex components lead to distinct disease entitites remains unresolved. In this study, authors establish the first Smarcb1 mutant mouse model with multiple brain abnormalities recapitulating human Coffin–Siris syndrome and show that one prominent midline abnormality, corpus callosum agenesis, is due to midline glia aberrations.
Collapse
Affiliation(s)
- Alina Filatova
- Stem Cell and Developmental Biology, Technical University Darmstadt, Darmstadt, 64287, Germany
| | - Linda K Rey
- Institute of Human Genetics, Medical Faculty, Heinrich Heine University, Düsseldorf, 40225, Germany
| | - Marion B Lechler
- Stem Cell and Developmental Biology, Technical University Darmstadt, Darmstadt, 64287, Germany
| | - Jörg Schaper
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University, Düsseldorf, 40225, Germany
| | - Maja Hempel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Renata Posmyk
- Podlaskie Medical Centre "GENETICS" Bialystok and Department of Perinatology and Obstetrics, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Krzysztof Szczaluba
- Department of Medical Genetics, Medical University Warsaw, Warsaw, 02-106, Poland
| | - Gijs W E Santen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2333 ZA, Netherlands
| | - Dagmar Wieczorek
- Institute of Human Genetics, Medical Faculty, Heinrich Heine University, Düsseldorf, 40225, Germany
| | - Ulrike A Nuber
- Stem Cell and Developmental Biology, Technical University Darmstadt, Darmstadt, 64287, Germany.
| |
Collapse
|
42
|
FGF Signaling Directs the Cell Fate Switch from Neurons to Astrocytes in the Developing Mouse Cerebral Cortex. J Neurosci 2019; 39:6081-6094. [PMID: 31175212 DOI: 10.1523/jneurosci.2195-18.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 05/09/2019] [Accepted: 05/17/2019] [Indexed: 11/21/2022] Open
Abstract
During mammalian neocortical development, neural precursor cells generate neurons first and astrocytes later. The cell fate switch from neurons to astrocytes is a key process generating proper numbers of neurons and astrocytes. Although the intracellular mechanisms regulating this cell fate switch have been well characterized, extracellular regulators are still largely unknown. Here, we uncovered that fibroblast growth factor (FGF) regulates the cell fate switch from neurons to astrocytes in the developing cerebral cortex using mice of both sexes. We found that the FGF signaling pathway is activated in radial glial cells of the ventricular zone at time points corresponding to the switch in cell fate. Our loss- and gain-of-function studies using in utero electroporation indicate that activation of FGF signaling is necessary and sufficient to change cell fates from neurons to astrocytes. We further found that the FGF-induced neuron-astrocyte cell fate switch is mediated by the MAPK pathway. These results indicate that FGF is a critical extracellular regulator of the cell fate switch from neurons to astrocytes in the mammalian cerebral cortex.SIGNIFICANCE STATEMENT Although the intracellular mechanisms regulating the neuron-astrocyte cell fate switch in the mammalian cerebral cortex during development have been well studied, their upstream extracellular regulators remain unknown. By using in utero electroporation, our study provides in vivo data showing that activation of FGF signaling is necessary and sufficient for changing cell fates from neurons to astrocytes. Manipulation of FGF signaling activity led to drastic changes in the numbers of neurons and astrocytes. These results indicate that FGF is a key extracellular regulator determining the numbers of neurons and astrocytes in the mammalian cerebral cortex, and is indispensable for the establishment of appropriate neural circuitry.
Collapse
|
43
|
A novel role for NFIA in restoring radiosensitivity in radioresistant NSCLC cells by downregulating the AKT and ERK pathways. Biochem Biophys Res Commun 2019; 515:558-564. [PMID: 31178144 DOI: 10.1016/j.bbrc.2019.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 06/02/2019] [Indexed: 12/27/2022]
Abstract
Radioresistance remains the most challenging issue leading to radiotherapy failure in the treatment of non-small cell lung cancer (NSCLC). The nuclear factor IA (NFIA) is associated with tumor response to treatments in many cancers, but its role in NSCLC radioresistance remains unclear. Here, we established two radioresistant NSCLC cell lines, H226R and H460R, by dose-gradient irradiation to investigate the function of NFIA in NSCLC radioresistance. The results showed a dramatically reduced expression of NFIA in radioresistant cells accompanied with elevated phosphorylation of AKT and ERK, when compared with their parental cells. Overexpression of NFIA restored the sensitivity of radioresistant cells to radiation through increased ionizing radiation (IR)-induced apoptosis and DNA damage by downregulating p-AKT and p-ERK, whereas knockdown of NFIA promoted radioresistance of the parental cells. Our findings suggested that NFIA enhanced cell radiosensitivity by downregulating p-AKT and p-ERK in NSCLC. Our study fills a gap in the field of NFIA and radioresistance, and establishes a mechanistic foundation to improve radiotherapy efficiency in NSCLC patients.
Collapse
|
44
|
Abstract
Normal brain development is best evaluated by MR imaging in the fetal and pediatric patient. As the developing brain grows, myelinates, and sulcates rapidly, understanding the normal appearance of the brain throughout development is critical. The fetal brain can be evaluated by MR imaging after 16 weeks gestational age, both morphologically and biometrically. Sulcation of the fetal brain lags behind premature neonates of equivalent gestational age. Sensory axons generally myelinate before motor axons with central to peripheral and dorsal to ventral myelination gradients. By 2 years of age, the brain has a near adult appearance by conventional anatomic MR imaging.
Collapse
Affiliation(s)
- Matthew J Barkovich
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, 505 Parnassus Avenue, Room S257, San Francisco, CA 94143-0628, USA
| | - Anthony James Barkovich
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, 505 Parnassus Avenue, Room S257, San Francisco, CA 94143-0628, USA.
| |
Collapse
|
45
|
Abstract
The corpus callosum is the largest of the 3 telencephalic commissures in eutherian (placental) mammals. Although the anterior commissure, and the hippocampal commissure before being pushed dorsally by the expanding frontal lobes, cross through the lamina reuniens (upper part of the lamina terminalis), the callosal fibers need a transient interhemispheric cellular bridge to cross. This review describes the molecular pathways that initiate the specification of the cells comprising this bridge, the specification of the callosal neurons, and the repulsive and attractive guidance molecules that convey the callosal axons toward, across, and away from the midline to connect with their targets.
Collapse
|
46
|
Heparan Sulfate Sulfation by Hs2st Restricts Astroglial Precursor Somal Translocation in Developing Mouse Forebrain by a Non-Cell-Autonomous Mechanism. J Neurosci 2019; 39:1386-1404. [PMID: 30617207 PMCID: PMC6381258 DOI: 10.1523/jneurosci.1747-17.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 12/27/2022] Open
Abstract
Heparan sulfate (HS) is a cell surface and extracellular matrix carbohydrate extensively modified by differential sulfation. HS interacts physically with canonical fibroblast growth factor (FGF) proteins that signal through the extracellular signal regulated kinase (ERK)/mitogen activated protein kinase (MAPK) pathway. At the embryonic mouse telencephalic midline, FGF/ERK signaling drives astroglial precursor somal translocation from the ventricular zone of the corticoseptal boundary (CSB) to the induseum griseum (IG), producing a focus of Slit2-expressing astroglial guidepost cells essential for interhemispheric corpus callosum (CC) axon navigation. Here, we investigated the cell and molecular function of a specific form of HS sulfation, 2-O HS sulfation catalyzed by the enzyme Hs2st, in midline astroglial development and in regulating FGF protein levels and interaction with HS. Hs2st−/− embryos of either sex exhibit a grossly enlarged IG due to precocious astroglial translocation and conditional Hs2st mutagenesis and ex vivo culture experiments show that Hs2st is not required cell autonomously by CC axons or by the IG astroglial cell lineage, but rather acts non-cell autonomously to suppress the transmission of translocation signals to astroglial precursors. Rescue of the Hs2st−/− astroglial translocation phenotype by pharmacologically inhibiting FGF signaling shows that the normal role of Hs2st is to suppress FGF-mediated astroglial translocation. We demonstrate a selective action of Hs2st on FGF protein by showing that Hs2st (but not Hs6st1) normally suppresses the levels of Fgf17 protein in the CSB region in vivo and use a biochemical assay to show that Hs2st (but not Hs6st1) facilitates a physical interaction between the Fgf17 protein and HS. SIGNIFICANCE STATEMENT We report a novel non-cell-autonomous mechanism regulating cell signaling in developing brain. Using the developing mouse telencephalic midline as an exemplar, we show that the specific sulfation modification of the cell surface and extracellular carbohydrate heparan sulfate (HS) performed by Hs2st suppresses the supply of translocation signals to astroglial precursors by a non-cell-autonomous mechanism. We further show that Hs2st modification selectively facilitates a physical interaction between Fgf17 and HS and suppresses Fgf17 protein levels in vivo, strongly suggesting that Hs2st acts selectively on Fgf17 signaling. HS interacts with many signaling proteins potentially encoding numerous selective interactions important in development and disease, so this class of mechanism may apply more broadly to other biological systems.
Collapse
|
47
|
Transcriptional control of long-range cortical projections. Curr Opin Neurobiol 2018; 53:57-65. [DOI: 10.1016/j.conb.2018.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/23/2018] [Indexed: 12/11/2022]
|
48
|
Schanze I, Bunt J, Lim JWC, Schanze D, Dean RJ, Alders M, Blanchet P, Attié-Bitach T, Berland S, Boogert S, Boppudi S, Bridges CJ, Cho MT, Dobyns WB, Donnai D, Douglas J, Earl DL, Edwards TJ, Faivre L, Fregeau B, Genevieve D, Gérard M, Gatinois V, Holder-Espinasse M, Huth SF, Izumi K, Kerr B, Lacaze E, Lakeman P, Mahida S, Mirzaa GM, Morgan SM, Nowak C, Peeters H, Petit F, Pilz DT, Puechberty J, Reinstein E, Rivière JB, Santani AB, Schneider A, Sherr EH, Smith-Hicks C, Wieland I, Zackai E, Zhao X, Gronostajski RM, Zenker M, Richards LJ. NFIB Haploinsufficiency Is Associated with Intellectual Disability and Macrocephaly. Am J Hum Genet 2018; 103:752-768. [PMID: 30388402 PMCID: PMC6218805 DOI: 10.1016/j.ajhg.2018.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022] Open
Abstract
The nuclear factor I (NFI) family of transcription factors play an important role in normal development of multiple organs. Three NFI family members are highly expressed in the brain, and deletions or sequence variants in two of these, NFIA and NFIX, have been associated with intellectual disability (ID) and brain malformations. NFIB, however, has not previously been implicated in human disease. Here, we present a cohort of 18 individuals with mild ID and behavioral issues who are haploinsufficient for NFIB. Ten individuals harbored overlapping microdeletions of the chromosomal 9p23-p22.2 region, ranging in size from 225 kb to 4.3 Mb. Five additional subjects had point sequence variations creating a premature termination codon, and three subjects harbored single-nucleotide variations resulting in an inactive protein as determined using an in vitro reporter assay. All individuals presented with additional variable neurodevelopmental phenotypes, including muscular hypotonia, motor and speech delay, attention deficit disorder, autism spectrum disorder, and behavioral abnormalities. While structural brain anomalies, including dysgenesis of corpus callosum, were variable, individuals most frequently presented with macrocephaly. To determine whether macrocephaly could be a functional consequence of NFIB disruption, we analyzed a cortex-specific Nfib conditional knockout mouse model, which is postnatally viable. Utilizing magnetic resonance imaging and histology, we demonstrate that Nfib conditional knockout mice have enlargement of the cerebral cortex but preservation of overall brain structure and interhemispheric connectivity. Based on our findings, we propose that haploinsufficiency of NFIB causes ID with macrocephaly.
Collapse
Affiliation(s)
- Ina Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Jens Bunt
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Jonathan W C Lim
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Denny Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Ryan J Dean
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Marielle Alders
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Patricia Blanchet
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Tania Attié-Bitach
- INSERM U1163, Laboratory of Embryology and Genetics of Congenital Malformations, Paris Descartes University, Sorbonne Paris Cité and Imagine Institute, Paris 75015, France
| | - Siren Berland
- Department of Medical Genetics, Haukeland University Hospital, Bergen 5021, Norway
| | - Steven Boogert
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Sangamitra Boppudi
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Caitlin J Bridges
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | | | - William B Dobyns
- Department of Pediatrics (Genetics), University of Washington and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Dian Donnai
- Manchester Centre for Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust; Division of Evolution and Genomic Sciences School of Biological Sciences, and University of Manchester, Manchester M13 9WL, UK
| | - Jessica Douglas
- Boston Children's Hospital - The Feingold Center, Waltham, MA 02115, USA
| | - Dawn L Earl
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Timothy J Edwards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; The Faculty of Medicine Brisbane, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Laurence Faivre
- UMR1231, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon 21079, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est et FHU TRANSLAD, Centre Hospitalier Universitaire Dijon, Dijon 21079, France
| | - Brieana Fregeau
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David Genevieve
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Marion Gérard
- Service de Génétique, CHU de Caen - Hôpital Clémenceau, Caen Cedex 14000, France
| | - Vincent Gatinois
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Muriel Holder-Espinasse
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille 59000, France; Department of Clinical Genetics, Guy's Hospital, London SE1 9RT, UK
| | - Samuel F Huth
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kosuke Izumi
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bronwyn Kerr
- Manchester Centre for Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust; Division of Evolution and Genomic Sciences School of Biological Sciences, and University of Manchester, Manchester M13 9WL, UK
| | - Elodie Lacaze
- Department of genetics, Le Havre Hospital, 76600 Le Havre, France
| | - Phillis Lakeman
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Sonal Mahida
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Ghayda M Mirzaa
- Department of Pediatrics (Genetics), University of Washington and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Sian M Morgan
- All Wales Genetics Laboratory, Institute of Medical Genetics, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Catherine Nowak
- Boston Children's Hospital - The Feingold Center, Waltham, MA 02115, USA
| | - Hilde Peeters
- Center for Human Genetics, University Hospital Leuven, KU Leuven, Leuven 3000, Belgium
| | - Florence Petit
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille 59000, France
| | - Daniela T Pilz
- West of Scotland Genetics Service, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Jacques Puechberty
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Eyal Reinstein
- Medical Genetics Institute, Meir Medical Center, Kfar-Saba 4428164, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Jean-Baptiste Rivière
- UMR1231, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon 21079, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est et FHU TRANSLAD, Centre Hospitalier Universitaire Dijon, Dijon 21079, France; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Avni B Santani
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anouck Schneider
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Elliott H Sherr
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Ilse Wieland
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Elaine Zackai
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaonan Zhao
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany.
| | - Linda J Richards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, The Faculty of Medicine Brisbane, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
49
|
Suárez R, Paolino A, Fenlon LR, Morcom LR, Kozulin P, Kurniawan ND, Richards LJ. A pan-mammalian map of interhemispheric brain connections predates the evolution of the corpus callosum. Proc Natl Acad Sci U S A 2018; 115:9622-9627. [PMID: 30181276 PMCID: PMC6156618 DOI: 10.1073/pnas.1808262115] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The brain of mammals differs from that of all other vertebrates, in having a six-layered neocortex that is extensively interconnected within and between hemispheres. Interhemispheric connections are conveyed through the anterior commissure in egg-laying monotremes and marsupials, whereas eutherians evolved a separate commissural tract, the corpus callosum. Although the pattern of interhemispheric connectivity via the corpus callosum is broadly shared across eutherian species, it is not known whether this pattern arose as a consequence of callosal evolution or instead corresponds to a more ancient feature of mammalian brain organization. Here we show that, despite cortical axons using an ancestral commissural route, monotremes and marsupials share features of interhemispheric connectivity with eutherians that likely predate the origin of the corpus callosum. Based on ex vivo magnetic resonance imaging and tractography, we found that connections through the anterior commissure in both fat-tailed dunnarts (Marsupialia) and duck-billed platypus (Monotremata) are spatially segregated according to cortical area topography. Moreover, cell-resolution retrograde and anterograde interhemispheric circuit mapping in dunnarts revealed several features shared with callosal circuits of eutherians. These include the layered organization of commissural neurons and terminals, a broad map of connections between similar (homotopic) regions of each hemisphere, and regions connected to different areas (heterotopic), including hyperconnected hubs along the medial and lateral borders of the cortex, such as the cingulate/motor cortex and claustrum/insula. We therefore propose that an interhemispheric connectome originated in early mammalian ancestors, predating the evolution of the corpus callosum. Because these features have been conserved throughout mammalian evolution, they likely represent key aspects of neocortical organization.
Collapse
Affiliation(s)
- Rodrigo Suárez
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4070, Australia;
| | - Annalisa Paolino
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4070, Australia
| | - Laura R Fenlon
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4070, Australia
| | - Laura R Morcom
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4070, Australia
| | - Peter Kozulin
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4070, Australia
| | - Nyoman D Kurniawan
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4070, Australia
| | - Linda J Richards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4070, Australia;
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4070, Australia
| |
Collapse
|
50
|
Bunt J, Osinski JM, Lim JW, Vidovic D, Ye Y, Zalucki O, O'Connor TR, Harris L, Gronostajski RM, Richards LJ, Piper M. Combined allelic dosage of Nfia and Nfib regulates cortical development. Brain Neurosci Adv 2017; 1:2398212817739433. [PMID: 32166136 PMCID: PMC7058261 DOI: 10.1177/2398212817739433] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/01/2017] [Indexed: 12/02/2022] Open
Abstract
Background: Nuclear factor I family members nuclear factor I A and nuclear factor I B play important roles during cerebral cortical development. Nuclear factor I A and nuclear factor I B regulate similar biological processes, as their expression patterns, regulation of target genes and individual knockout phenotypes overlap. We hypothesised that the combined allelic loss of Nfia and Nfib would culminate in more severe defects in the cerebral cortex than loss of a single member. Methods: We combined immunofluorescence, co-immunoprecipitation, gene expression analysis and immunohistochemistry on knockout mouse models to investigate whether nuclear factor I A and nuclear factor I B function similarly and whether increasing allelic loss of Nfia and Nfib caused a more severe phenotype. Results: We determined that the biological functions of nuclear factor I A and nuclear factor I B overlap during early cortical development. These proteins are co-expressed and can form heterodimers in vivo. Differentially regulated genes that are shared between Nfia and Nfib knockout mice are highly enriched for nuclear factor I binding sites in their promoters and are associated with neurodevelopment. We found that compound heterozygous deletion of both genes resulted in a cortical phenotype similar to that of single homozygous Nfia or Nfib knockout embryos. This was characterised by retention of the interhemispheric fissure, dysgenesis of the corpus callosum and a malformed dentate gyrus. Double homozygous knockout of Nfia and Nfib resulted in a more severe phenotype, with increased ventricular enlargement and decreased numbers of differentiated glia and neurons. Conclusion: In the developing cerebral cortex, nuclear factor I A and nuclear factor I B share similar biological functions and function additively, as the combined allelic loss of these genes directly correlates with the severity of the developmental brain phenotype.
Collapse
Affiliation(s)
- Jens Bunt
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jason M Osinski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Jonathan Wc Lim
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Diana Vidovic
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Yunan Ye
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Oressia Zalucki
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy R O'Connor
- School of Chemical and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Lachlan Harris
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Linda J Richards
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.,The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Michael Piper
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.,The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|